Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Anesth Analg ; 129(5): 1414-1421, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30044299

RESUMO

BACKGROUND: A major advancement in the field of analgesic pharmacology has been the development of G-protein-biased opioid agonists that display less respiratory depression than conventional drugs. It is uncertain, however, whether these new drugs cause less tolerance, hyperalgesia, and other maladaptations when administered repeatedly. METHODS: The archetypical µ-opioid receptor agonist morphine and, separately, the G-protein-biased µ-opioid receptor agonist oliceridine were administered to mice. These drugs were used in models of acute analgesia, analgesic tolerance, opioid-induced hyperalgesia, reward, and physical dependence. In addition, morphine and oliceridine were administered for 7 days after tibia fracture and pinning; mechanical allodynia and gait were followed for 3 weeks. Finally, the expression of toll-like receptor-4 and nacht domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NALP3) and interleukin-1ß mRNA were quantified in spinal tissue to measure surgical and drug effects on glia-related gene expression. RESULTS: We observed using the tail flick assay that oliceridine was a 4-fold more potent analgesic than morphine, but that oliceridine treatment caused less tolerance and opioid-induced hyperalgesia than morphine after 4 days of ascending-dose administration. Using similar analgesic doses, morphine caused reward behavior in the conditioned place preference assay while oliceridine did not. Physical dependence was, however, similar for the 2 drugs. Likewise, morphine appeared to more significantly impair the recovery of nociceptive sensitization and gait after tibial fracture and pinning than oliceridine. Furthermore, spinal cord toll-like receptor-4 levels 3 weeks after fracture were higher in fracture mice given morphine than those given oliceridine. CONCLUSIONS: Aside from reduced respiratory depression, G-protein-biased agonists such as oliceridine may reduce opioid maladaptations and enhance the quality of surgical recovery.


Assuntos
Receptores Opioides mu/agonistas , Compostos de Espiro/farmacologia , Tiofenos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Dor Pós-Operatória/tratamento farmacológico , Fraturas da Tíbia/fisiopatologia , Receptor 4 Toll-Like/análise
2.
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
3.
BMC Genomics ; 17: 313, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27129385

RESUMO

BACKGROUND: Opioids are a mainstay for the treatment of chronic pain. Unfortunately, therapy-limiting maladaptations such as loss of treatment effect (tolerance), and paradoxical opioid-induced hyperalgesia (OIH) can occur. The objective of this study was to identify genes responsible for opioid tolerance and OIH. RESULTS: These studies used a well-established model of ascending morphine administration to induce tolerance, OIH and other opioid maladaptations in 23 strains of inbred mice. Genome-wide computational genetic mapping was then applied to the data in combination with a false discovery rate filter. Transgenic mice, gene expression experiments and immunoprecipitation assays were used to confirm the functional roles of the most strongly linked gene. The behavioral data processed using computational genetic mapping and false discovery rate filtering provided several strongly linked biologically plausible gene associations. The strongest of these was the highly polymorphic Mpdz gene coding for the post-synaptic scaffolding protein Mpdz/MUPP1. Heterozygous Mpdz +/- mice displayed reduced opioid tolerance and OIH. Mpdz gene expression and Mpdz/MUPP1 protein levels were lower in the spinal cords of low-adapting 129S1/Svlm mice than in high-adapting C57BL/6 mice. Morphine did not alter Mpdz expression levels. In addition, association of Mpdz/MUPP1 with its known binding partner CaMKII did not differ between these high- and low-adapting strains. CONCLUSIONS: The degrees of maladaptive changes in response to repeated administration of morphine vary greatly across inbred strains of mice. Variants of the multiple PDZ domain gene Mpdz may contribute to the observed inter-strain variability in tolerance and OIH by virtue of changes in the level of their expression.


Assuntos
Proteínas de Transporte/genética , Tolerância a Medicamentos/genética , Hiperalgesia/genética , Morfina/efeitos adversos , Domínios PDZ , Analgésicos Opioides/efeitos adversos , Animais , Mapeamento Cromossômico , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes , Haplótipos , Hiperalgesia/induzido quimicamente , Masculino , Proteínas de Membrana , Camundongos Endogâmicos , Camundongos Transgênicos , Dependência de Morfina/genética , Polimorfismo de Nucleotídeo Único
4.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27094549

RESUMO

BACKGROUND: Opioids have become the mainstay for treatment of moderate to severe pain and are commonly used to treat surgical pain. While opioid administration has been shown to cause opioid-induced hyperalgesia and tolerance, interactions between opioid administration and surgery with respect to these problematic adaptations have scarcely been addressed. Accumulating evidence suggests opioids and nociceptive signaling may converge on epigenetic mechanisms in spinal cord to enhance or prolong neuroplastic changes. Epigenetic regulation of Bdnf (brain-derived neurotrophic factor) and Pdyn (prodynorphin) genes may be involved. RESULTS: Four days of ascending doses of morphine treatment caused opioid-induced hyperalgesia and reduced opioid analgesic efficacy in mice. Both opioid-induced hyperalgesia and the reduced opioid analgesic efficacy were enhanced in mice that received hindpaw incisions. The expression of Bdnf and Pdyn (qPCR) was increased after morphine treatment and incision. Chromatin immunoprecipitation assays demonstrated that the Pdyn and Bdnf promoters were more strongly associated with acetylated H3K9 after morphine plus incision than in the morphine or incision alone groups. Selective tropomyosin-related kinase B (ANA-12) and κ-opioid receptor (nor-binaltorphimine) antagonists were administered intrathecally, both reduced hyperalgesia one or three days after surgery. Administration of ANA-12 or nor-binaltorphimine attenuated the decreased morphine analgesic efficacy on day 1, but only nor-binaltorphimine was effective on day 3 after incision in opioid-exposed group. Coadministration of histone acetyltransferase inhibitor anacardic acid daily with morphine blocked the development of opioid-induced hyperalgesia and attenuated incision-enhanced hyperalgesia in morphine-treated mice. Anacardic acid had similar effects on analgesic tolerance, showing the involvement of histone acetylation in the interactions detected. CONCLUSIONS: Spinal epigenetic changes involving Bdnf and Pdyn may contribute to the enhanced postoperative nociceptive sensitization and analgesic tolerance observed after continuous opioid exposure. Treatments blocking the epigenetically mediated up-regulation of these genes or administration of TrkB or κ-opioid receptor antagonists may improve the clinical utility of opioids, particularly after surgery.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Tolerância a Medicamentos , Epigênese Genética/efeitos dos fármacos , Dor Pós-Operatória/tratamento farmacológico , Dor Pós-Operatória/genética , Medula Espinal/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dinorfinas/metabolismo , Histona Acetiltransferases/antagonistas & inibidores , Histona Acetiltransferases/metabolismo , Hiperalgesia/complicações , Hiperalgesia/genética , Hiperalgesia/patologia , Masculino , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/cirurgia
5.
BMC Genomics ; 15: 345, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24884839

RESUMO

BACKGROUND: Opioids are the cornerstone of treatment for moderate to severe pain, but chronic use leads to maladaptations that include: tolerance, dependence and opioid-induced hyperalgesia (OIH). These responses limit the utility of opioids, as well as our ability to control chronic pain. Despite decades of research, we have no therapies or proven strategies to overcome this problem. However, murine haplotype based computational genetic mapping and a SNP data base generated from analysis of whole-genome sequence data (whole-genome HBCGM), provides a hypothesis-free method for discovering novel genes affecting opioid maladaptive responses. RESULTS: Whole genome-HBCGM was used to analyze phenotypic data on morphine-induced tolerance, dependence and hyperalgesia obtained from 23 inbred strains. The robustness of the genetic mapping results was analyzed using strain subsets. In addition, the results of analyzing all of the opioid-related traits together were examined. To characterize the functional role of the leading candidate gene, we analyzed transgenic animals, mRNA and protein expression in behaviorally divergent mouse strains, and immunohistochemistry in spinal cord tissue. Our mapping procedure identified the allelic pattern within the netrin-1 receptor gene (Dcc) as most robustly associated with OIH, and it was also strongly associated with the combination of the other maladaptive opioid traits analyzed. Adult mice heterozygous for the Dcc gene had significantly less tendency to develop OIH, become tolerant or show evidence of dependence after chronic exposure to morphine. The difference in opiate responses was shown not to be due to basal or morphine-stimulated differences in the level of Dcc expression in spinal cord tissue, and was not associated with nociceptive neurochemical or anatomical alterations in the spinal cord or dorsal root ganglia in adult animals. CONCLUSIONS: Whole-genome HBCGM is a powerful tool for identifying genes affecting biomedical traits such as opioid maladaptations. We demonstrate that Dcc affects tolerance, dependence and OIH after chronic opioid exposure, though not through simple differences in expression in the adult spinal cord.


Assuntos
Hiperalgesia/induzido quimicamente , Morfina/administração & dosagem , Receptores de Superfície Celular/genética , Animais , Comportamento Animal/efeitos dos fármacos , Mapeamento Cromossômico , Bases de Dados Factuais , Tolerância a Medicamentos , Genoma , Haplótipos , Heterozigoto , Hiperalgesia/genética , Hiperalgesia/patologia , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Morfina/efeitos adversos , Morfina/farmacologia , Receptores de Netrina , Proteínas/metabolismo , RNA/metabolismo , Receptores de Superfície Celular/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
6.
Mol Pain ; 10: 59, 2014 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-25217253

RESUMO

BACKGROUND: The long term use of opioids for the treatment of pain leads to a group of maladaptations which includes opioid-induced hyperalgesia (OIH). OIH typically resolves within few days after cessation of morphine treatment in mice but is prolonged for weeks if histone deacetylase (HDAC) activity is inhibited during opioid treatment. The present work seeks to identify gene targets supporting the epigenetic effects responsible for OIH prolongation. RESULTS: Mice were treated with morphine according to an ascending dose protocol. Some mice also received the selective HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) additionally. Chronic morphine treatment with simultaneous HDAC inhibition enhanced OIH, and several spinal cord genes were up-regulated. The expression of Bdnf (Brain-derived neurotrophic factor) and Pdyn (Prodynorphin) were most closely related to the observed behavioral changes. ChIP (Chromatin immuoprecipation) assays demonstrated that promoter regions of Pdyn and Bdnf were strongly associated with aceH3K9 (Acetylated histone H3 Lysine9) after morphine and SAHA treatment. Furthermore, morphine treatment caused an increase in spinal BDNF and dynorphin levels, and these levels were further increased in SAHA treated mice. The selective TrkB (tropomyosin-receptor-kinase) antagonist ANA-12 reduced OIH when given one or seven days after cessation of morphine. Treatment with the selective kappa opioid receptor antagonist nor-BNI also reduced established OIH. The co-administration of either receptor antagonist agent daily with morphine resulted in attenuation of hyperalgesia present one day after cessation of treatment. Additionally, repeated morphine exposure induced a rise in BDNF expression that was associated with an increased number of BDNF+ cells in the spinal cord dorsal horn, showing strong co-localization with aceH3K9 in neuronal cells. Lastly, spinal application of low dose BDNF or Dynorphin A after resolution of OIH produced mechanical hypersensitivity, with no effect in controls. CONCLUSIONS: The present study identified two genes whose expression is regulated by epigenetic mechanisms during morphine exposure. Treatments aimed at preventing the acetylation of histones or blocking BDNF and dynorphin signaling may reduce OIH and improve long-term pain using opioids.


Assuntos
Analgésicos Opioides/toxicidade , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hiperalgesia/induzido quimicamente , Hiperalgesia/patologia , Morfina/toxicidade , Medula Espinal/metabolismo , Animais , Antineoplásicos/administração & dosagem , Azepinas/administração & dosagem , Benzamidas/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Dinorfinas/administração & dosagem , Dinorfinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Ácidos Hidroxâmicos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/administração & dosagem , Medição da Dor/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Vorinostat
7.
Anesth Analg ; 118(6): 1336-44, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24755847

RESUMO

BACKGROUND: Acute pain after surgery remains moderate to severe for 20% to 30% of patients despite advancements in the use of opioids, adjuvant drugs, and regional anesthesia. Depending on the type of surgery, 10% to 50% of patients experience persistent pain postoperatively, and there are no established methods for its prevention. Curcumin (diferuloylmethane) is one of the phenolic constituents of turmeric that has been used in Eastern traditional medicine as an antiseptic, antioxidant, anti-inflammatory, and analgesic agent. It may be effective for treating postoperative pain. METHODS: We used the hindpaw incision model with C57BL/6 mice. Sensitization to mechanical and thermal stimuli as well as effects on edema and temperature were measured up to 7 days after surgery. Spontaneous pain after incision was assessed by using conditioned place preference (CPP), and alterations in gait function were assessed using multiparameter digital gait analysis. RESULTS: Curcumin (50 mg/kg) significantly reduced the intensity of mechanical and heat sensitization after hindpaw incision in mice. No effects of curcumin on baseline nociceptive thresholds were observed. Curcumin also reduced hindpaw swelling after incision, suggesting an anti-inflammatory effect. In addition, perioperative curcumin treatment attenuated hyperalgesic priming due to incision when mice were subsequently challenged with hindpaw prostaglandin E2 application. Furthermore, while vehicle-treated mice had evidence of spontaneous pain 48 hours after incision in the CPP paradigm, no evidence of ongoing pain was observed in the mice treated with curcumin. Likewise, hindpaw incision caused changes in several gait-related indices, but most of these were normalized in the curcumin-treated animals. The peri-incisional levels of several pronociceptive immune mediators including interleukin (IL)-1ß, IL-6, tumor necrosis factor α, and macrophage inflammatory protein-1α were either not reduced or were even augmented 1 and 3 days after incision in curcumin-treated mice. The anti-inflammatory cytokine IL-10 was unchanged, while transforming growth factor-ß levels were enhanced under the same conditions. CONCLUSIONS: Our studies suggest that curcumin treatment is effective in alleviating incision-induced inflammation, nociceptive sensitization, spontaneous pain, and functional gait abnormalities. Augmented transforming growth factor-ß production provides one possible mechanism. These preclinical findings demonstrate curcumin's potential as a preventative strategy in postoperative pain treatment.


Assuntos
Dor Aguda/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Curcumina/farmacologia , Dor Pós-Operatória/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Animais , Fenômenos Biomecânicos , Temperatura Corporal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Citocinas/biossíntese , Edema/patologia , Edema/prevenção & controle , Traumatismos do Pé/complicações , Traumatismos do Pé/tratamento farmacológico , Marcha/efeitos dos fármacos , Membro Posterior/lesões , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medição da Dor/efeitos dos fármacos , Resultado do Tratamento
8.
Anesthesiology ; 119(5): 1198-208, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23756451

RESUMO

BACKGROUND: The regulation of gene expression in nociceptive pathways contributes to the induction and maintenance of pain sensitization. Histone acetylation is a key epigenetic mechanism controlling chromatin structure and gene expression. Chemokine CC motif receptor 2 (CXCR2) is a proinflammatory receptor implicated in neuropathic and inflammatory pain and is known to be regulated by histone acetylation in some settings. The authors sought to investigate the role of histone acetylation on spinal CXCR2 signaling after incision. METHODS: Groups of 5-8 mice underwent hind paw incision. Suberoylanilide hydroxamic acid and anacardic acid were used to inhibit histone deacetylase and histone acetyltransferase, respectively. Behavioral measures of thermal and mechanical sensitization as well as hyperalgesic priming were used. Both message RNA quantification and chromatin immunoprecipitation analysis were used to study the regulation of CXCR2 and ligand expression. Finally, the selective CXCR2 antagonist SB225002 was administered intrathecally to reveal the function of spinal CXCR2 receptors after hind paw incision. RESULTS: Suberoylanilide hydroxamic acid significantly exacerbated mechanical sensitization after incision. Conversely, anacardic acid reduced incisional sensitization and also attenuated incision-induced hyperalgesic priming. Overall, acetylated histone H3 at lysine 9 was increased in spinal cord tissues after incision, and enhanced association of acetylated histone H3 at lysine 9 with the promoter regions of CXCR2 and keratinocyte-derived chemokine (CXCL1) was observed as well. Blocking CXCR2 reversed mechanical hypersensitivity after hind paw incision. CONCLUSIONS: Histone modification is an important epigenetic mechanism regulating incision-induced nociceptive sensitization. The spinal CXCR2 signaling pathway is one epigenetically regulated pathway controlling early and latent sensitization after incision.


Assuntos
Epigênese Genética/fisiologia , Hiperalgesia/genética , Período Intraoperatório , Nociceptividade/fisiologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Medula Espinal/fisiopatologia , Ácidos Anacárdicos/administração & dosagem , Ácidos Anacárdicos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Western Blotting , Imunoprecipitação da Cromatina , Dinoprostona/administração & dosagem , Dinoprostona/farmacologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Histona Acetiltransferases/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Hiperalgesia/etiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medição da Dor/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Estimulação Física , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina-8B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Vorinostat
9.
Anesthesiology ; 117(3): 626-38, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22846677

RESUMO

BACKGROUND: After incision keratinocytes in the epidermis become activated to produce a range of pain-related mediators. microRNA 203 (miR-203) is known to be involved in keratinocyte growth, differentiation, and skin inflammation. We hypothesized that one or more of these mediators might be under the control of miR-203. METHODS: The expression of miR-203 and its target gene, phospholipase A2 activating protein (PLAA), were examined after hind paw incision in mice. We investigated the local effect of intraplantar PLAA peptide injection in normal mice and the effects of a selective secretory phospholipase A2 inhibitor (HK064) on PLAA or incision-induced mechanical allodynia. Last, we investigated the role of substance P signaling in regulating miR-203 and PLAA expression in vitro and in vivo. RESULTS: Levels of miR-203 were strongly down-regulated in keratinocytes after incision. Informatics-based approaches identified PLAA as a likely candidate for regulation by miR-203. PLAA caused mechanical allodynia and conditioned place aversion but not thermal sensitization. HK064 reduced mechanical allodynia after incision and after intraplantar injection of PLAA. Using preprotachykinin gene knockout mice or with neurokinin-1 selective antagonist LY303870 treatment, we observed that substance P-mediated signaling was also required for miR-203 and PLAA regulation after incision. Finally, using the rat epidermal keratinocyte cell line, we observed that a miR-203 mimic molecule could block the substance P-induced increase in PLAA expression observed under control conditions. CONCLUSIONS: miR-203 may regulate expression of the novel nociceptive mediator PLAA after incision. Furthermore, the regulation of miR-203 and PLAA levels is reliant upon intact substance P signaling.


Assuntos
MicroRNAs/fisiologia , Dor/fisiopatologia , Proteínas/fisiologia , Animais , Condicionamento Psicológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Precursores de Proteínas/fisiologia , Receptores da Neurocinina-1/fisiologia , Transdução de Sinais/fisiologia , Substância P/fisiologia , Taquicininas/fisiologia
10.
Anesthesiology ; 116(4): 882-95, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22343473

RESUMO

BACKGROUND: Patients with complex regional pain syndrome have increased tryptase in the skin of the affected extremity indicating mast cell (MC) accumulation and degranulation, processes known to be mediated by substance P (SP). The dysregulation of SP release from primary afferent neurons is characteristic of complex regional pain syndrome. The authors hypothesized that SP acting through the neurokinin-1 receptor results in mast cell accumulation, degranulation, and nociceptive sensitization in a rat model of complex regional pain syndrome. METHODS: Groups of 6-10 rats underwent tibia fracture and hind limb casting for 4 weeks, and the hind paw skin was harvested for histologic and immunohistochemical analysis. The effects of a selective neurokinin-1 receptor antagonist (LY303870) and of direct SP intraplantar injection were measured. Dermal MC degranulation induced by sciatic nerve stimulation and the effects of LY303870 on this process were investigated. Finally, the antinociceptive effects of acute and chronic treatment with a MC degranulator (48/80) were tested. RESULTS: The authors observed that fracture caused MC accumulation, activation, and degranulation, which were inhibited by LY303870; the percentage of MCs in close proximity to peptidergic nerve fibers increased after fracture; electrical stimulation caused MC activation and degranulation, which was blocked by LY303870; intraplantar SP-induced MC degranulation and acute administration of 48/80 caused MC degranulation and enhanced postfracture nociception, but MC-depleted animals showed less sensitization. CONCLUSIONS: These results indicate that facilitated peptidergic neuron-MC signaling after fracture can cause MC accumulation, activation, and degranulation in the injured limb, resulting in nociceptive sensitization.


Assuntos
Síndromes da Dor Regional Complexa/metabolismo , Modelos Animais de Doenças , Mastócitos/metabolismo , Nociceptividade/fisiologia , Substância P/fisiologia , Fraturas da Tíbia/metabolismo , Animais , Síndromes da Dor Regional Complexa/patologia , Indóis/farmacologia , Masculino , Mastócitos/efeitos dos fármacos , Antagonistas dos Receptores de Neurocinina-1 , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Piperidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/metabolismo
11.
Anesthesiology ; 114(5): 1180-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21368652

RESUMO

BACKGROUND: Opioid-induced hyperalgesia (OIH) and tolerance are challenging maladaptations associated with opioids in managing pain. Recent genetic studies and the existing literature suggest the 5-hydroxytryptamine type 3 (5-HT3) receptor participates in these phenomena. The location of the relevant receptor populations and the interactions between the 5-HT3 system and other systems controlling OIH and tolerance have not been explored, however. We hypothesized that 5-HT3 receptors modulate OIH and tolerance, and that this modulation involves the control of expression of multiple neurotransmitter and receptor systems. METHODS: C57BL/6 mice were exposed to a standardized 4-day morphine administration protocol. The 5-HT3 antagonist ondansetron was administered either during or after the conclusion of morphine administration. Mechanical testing was used to quantify OIH, and thermal tail-flick responses were used to measure morphine tolerance. In other experiments spinal cord and dorsal root ganglion tissues were harvested for analysis of messenger RNA concentrations by real-time polymerase chain reaction or immunochemistry analysis. RESULTS: The results showed that (1) systemic or intrathecal injection of ondansetron significantly prevented and reversed OIH, but not local intraplantar injection; (2) systemic or intrathecal injection of ondansetron prevented and reversed tolerance; and (3) ondansetron blocked morphine-induced increases of multiple genes relevant to OIH and tolerance in dorsal root ganglion and spinal cord. CONCLUSIONS: Morphine acts via a 5-HT3-dependent mechanism to support multiple maladaptations to the chronic administration of morphine. Furthermore, the use of 5-HT3 receptor antagonists may provide a new avenue to prevent or reverse OIH and tolerance associated with chronic opioid use.


Assuntos
Analgésicos Opioides/efeitos adversos , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Morfina/efeitos adversos , Ondansetron/administração & dosagem , Receptores 5-HT3 de Serotonina/metabolismo , Antagonistas da Serotonina/administração & dosagem , Animais , Tolerância a Medicamentos , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Anesthesiology ; 113(4): 945-56, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20823759

RESUMO

BACKGROUND: Surgical injury induces production and release of inflammatory mediators in the vicinity of the wound. They in turn trigger nociceptive signaling to produce hyperalgesia and pain. Interleukin-1ß plays a crucial role in this process. The mechanism regulating production of this cytokine after incision is, however, unknown. Caspase-1 is a key enzyme that cleaves prointerleukin-1ß to its active form. We hypothesized that caspase-1 is a crucial regulator of incisional interleukin-1ß levels, nociceptive sensitization, and inflammation. METHODS: These studies employed a mouse hind paw incisional model. Caspase-1 was blocked using the selective inhibitors Ac-YVAD-CMK and VRTXSD727. Nociceptive sensitization, edema, and hind paw warmth were followed in intact animals whereas caspase-1 activity, cytokine, and prostaglandin E2 levels were assessed in homogenized skin. Confocal microscopy was used to detect the expression of caspase-1 near the wounds. RESULTS: Analysis of enzyme activity demonstrated that caspase-1 activity was significantly increased in periincisional skin. Pretreatment with Ac-YVAD-CMK significantly reduced mechanical allodynia and thermal hyperalgesia. Repeated administration of this inhibitor produced robust analgesia, especially to mechanical stimulation. Administration of VRTXSD727 provided qualitatively similar results. Caspase-1 inhibition also reduced edema and the normally observed increase in paw warmth around the wound site. Correspondingly, caspase-1 inhibition significantly reduced interleukin-1ß as well as macrophage-inflammatory protein 1α, granulocyte colony-stimulating factor, and prostaglandin E2 levels near the wound. The expression of caspase-1 was primarily observed in keratinocytes in the epidermal layer and in neutrophils deeper in the wounds. CONCLUSIONS: The current study demonstrates that the inhibition of caspase-1 reduces postsurgical sensitization and inflammation, likely through a caspase-1/interleukin-1ß-dependent mechanism.


Assuntos
Caspase 1/fisiologia , Inflamação/patologia , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Animais , Biomarcadores , Temperatura Corporal/efeitos dos fármacos , Inibidores de Caspase , Quimiocina CCL3/metabolismo , Dinoprostona/metabolismo , Edema/patologia , Fator Estimulador de Colônias de Granulócitos/líquido cefalorraquidiano , Membro Posterior/patologia , Temperatura Alta , Hiperalgesia/fisiopatologia , Imuno-Histoquímica , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dor/psicologia , Medição da Dor , Limiar da Dor/fisiologia , Pele/metabolismo
13.
Sci Rep ; 9(1): 19500, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31863005

RESUMO

High rates of acute and chronic pain are associated with traumatic brain injury (TBI), but mechanisms responsible for the association remain elusive. Recent data suggest dysregulated descending pain modulation circuitry could be involved. Based on these and other observations, we hypothesized that serotonin (5-HT)-dependent activation of spinal CXC Motif Chemokine Receptor 2 (CXCR2) may support TBI-related nociceptive sensitization in a mouse model of mild TBI (mTBI). We observed that systemic 5-HT depletion with p-chlorophenylalanine attenuated mechanical hypersensitivity seen after mTBI. Likewise, selective spinal 5-HT fiber depletion with 5,7-dihydroxytryptamine (5,7-DHT) reduced hypersensitivity after mTBI. Consistent with a role for spinal 5-HT3 serotonin receptors, intrathecal ondansetron administration after TBI dose-dependently attenuated nociceptive sensitization. Also, selective CXCR2 antagonist SCH527123 treatment attenuated mechanical hypersensitivity after mTBI. Furthermore, spinal CXCL1 and CXCL2 mRNA and protein levels were increased after mTBI as were GFAP and IBA-1 markers. Spinal 5,7-DHT application reduced both chemokine expression and glial activation. Our results suggest dual pathways for nociceptive sensitization after mTBI, direct 5-HT effect through 5-HT3 receptors and indirectly through upregulation of chemokine signaling. Designing novel clinical interventions against either the 5-HT3 mediated component or chemokine pathway may be beneficial in treating pain frequently seen in patients after mTBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Receptores 5-HT1 de Serotonina/metabolismo , 5,7-Di-Hidroxitriptamina/farmacologia , Animais , Benzamidas/farmacologia , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Ciclobutanos/farmacologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fenclonina/farmacologia , Imuno-Histoquímica , Masculino , Camundongos , Ondansetron/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina-8B/antagonistas & inibidores , Receptores de Interleucina-8B/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo
14.
J Neurotrauma ; 35(13): 1495-1509, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29373948

RESUMO

Chronic pain is a common consequence of traumatic brain injury (TBI) that can increase the suffering of a patient and pose a significant challenge to rehabilitative efforts. Unfortunately, the mechanisms linking TBI to pain are poorly understood, and specific treatments for TBI-related pain are still lacking. Our laboratory has shown that TBI causes pain sensitization in areas distant to the site of primary injury, and that changes in spinal gene expression may underlie this sensitization. The aim of this study was to examine the roles that pain modulatory pathways descending from the brainstem play in pain after TBI. Deficiencies in one type of descending inhibition, diffuse noxious inhibitory control (DNIC), have been suggested to be responsible for the development of chronic pain by allowing excess and uncontrolled afferent nociceptive inputs. Here we expand our knowledge of pain after TBI in two ways: (1) by outlining the neuropathology in pain-related centers of the brain and spinal cord involved in DNIC using the rat lateral fluid percussion (LFP) model of TBI, and (2) by evaluating the effects of a potent histone acetyl transferase inhibitor, anacardic acid (AA), on LFP-induced pain behaviors and neuropathology when administered for several days after TBI. The results revealed that TBI induces transient mechanical allodynia and a chronic persistent loss of DNIC. Further, while short-term AA treatment can block acute nociceptive sensitization and some early neuropathological changes, this treatment neither prevented the loss of DNIC nor did it alter long-term neuropathological changes in the brain or spinal cord.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Tronco Encefálico/fisiopatologia , Dor Crônica/fisiopatologia , Hiperalgesia/fisiopatologia , Medula Espinal/fisiopatologia , Animais , Lesões Encefálicas Traumáticas/complicações , Dor Crônica/etiologia , Masculino , Ratos , Ratos Long-Evans , Transdução de Sinais/fisiologia
15.
Behav Brain Res ; 181(1): 118-26, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17498818

RESUMO

Adaptations to the chronic administration of opioids reduce the utility of these drugs in treating pain and support addiction. Recent genetics-based approaches have implicated the beta2 adrenergic receptor (beta2-AR) in controlling some of these responses. We do not know, however, whether this receptor can modulate tolerance, dependence or changes in gene expression caused by chronic opioid administration. For our studies we used C57BL/6 mice and beta2-AR knockout mice in the FVB background. Morphine dose-response relationships were established both prior to and after chronic morphine treatment. In some cases, the selective beta2-AR antagonist butoxamine was administered along with or after morphine. Physical dependence was assessed using naloxone-precipitated withdrawal. The expression of calcitonin gene related peptide (CGRP) and substance P (SP) were measured in spinal cord and dorsal root ganglion (DRG) tissues using both real-time PCR and enzyme-linked immunoassay (ELISA). Both the co-administration of butoxamine with morphine and the administration of butoxamine after chronic morphine reversed morphine tolerance. Morphine failed to cause tolerance in beta2-AR knockout mice. Physical dependence was reduced under the same circumstances. The chronic administration of butoxamine with morphine reduced or eliminated the normally observed up-regulation of CGRP and SP in spinal cord and DRG tissues. Our results suggest that the beta2-AR modulates both opioid tolerance and physical dependence. Activation of beta2-ARs appears to be required for some of the key neurochemical changes which characterize chronic opioid administration. Therefore, beta2-AR antagonists show some promise as agents to enhance chronic opioid analgesic therapy.


Assuntos
Tolerância a Medicamentos/fisiologia , Dependência de Morfina/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Antagonistas Adrenérgicos beta/farmacologia , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Animal , Butoxamina/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Gânglios Espinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/administração & dosagem , Dependência de Morfina/patologia , RNA Mensageiro/biossíntese , Receptores Adrenérgicos beta 2/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Medula Espinal/metabolismo
16.
IBRO Rep ; 2: 14-23, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30135929

RESUMO

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. In these studies we pursued the hypothesis that TBI pain sensitization is associated with histone acetylation in the rat lateral fluid percussion model. Some animals received hindpaw incisions in addition to TBI to mimic polytrauma. Neuropathological analysis of brain tissue from sham and TBI animals revealed evidence of bleeding, breakdown of the blood brain barrier, in the cortex, hippocampus, thalamus and other structures related to pain signal processing. Mechanical allodynia was measured in these animals for up to eight weeks post-injury. Inhibitors of histone acetyltransferase (HAT) and histone deacetylase (HDAC) were used to probe the role of histone acetylation in such pain processing. We followed serum markers including glial fibrillary acidic protein (GFAP), neuron-specific enolase 2 (NSE) myelin basic protein (MBP) and S100ß to gauge TBI injury severity. Our results showed that TBI caused mechanical allodynia in the hindpaws of the rats lasting several weeks. Hindpaws contralateral to TBI showed more rapid and profound sensitization than ipsilateral hindpaws. The inhibition of HAT using curcumin 50 mg/kg s.c reduced mechanical sensitization while the HDAC inhibitor suberoylanilide hydroxamic acid 50 mg/kg i.p. prolonged sensitization in the TBI rats. Immunohistochemical analyses of spinal cord tissue localized changes in the level of acetylation of the H3K9 histone mark to dorsal horn neurons. Taken together, these findings demonstrate that TBI induces sustained nociceptive sensitization, and changes in spinal neuronal histone proteins may play an important role.

17.
Pain ; 121(3): 232-240, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16516386

RESUMO

Opioids are commonly used in the treatment of moderate to severe pain. However, their chronic use is limited by analgesic tolerance and physical dependence. Few studies have examined how chronic pain affects the development of tolerance or dependence, and essentially no studies have looked at the role of both genetics and pain together. For these studies we used 12 strains of inbred mice. Groups of mice from each strain were tested at baseline for morphine analgesic sensitivity, mechanical nociceptive threshold, and thermal nociceptive threshold. Mice were then given morphine in a 4-day escalating morphine administration paradigm followed by reassessment of the morphine dose-response relationship. Finally, physical dependence was measured by administering naloxone. Parallel groups of mice underwent hind paw injection of complete Freund's adjuvant (CFA) to induce chronic hind paw inflammation 7 days prior to the beginning of testing. The data showed that CFA treatment tended to lower baseline ED(50) values for morphine and enhanced the degree of analgesic tolerance observed after 4 days of morphine treatment. In addition, the degree of jumping behavior indicative of physical dependence was often altered if mice had been treated with CFA. The influence of background strain was substantial for all traits measured. In silico haplotypic mapping of the tolerance and physical dependence data demonstrated that CFA pretreatment altered the pattern of the predicted associations and greatly reduced their statistical significance. We conclude that chronic inflammatory pain and genetics interact to modulate the analgesic potency of morphine, tolerance, and physical dependence.


Assuntos
Analgésicos Opioides/farmacologia , Predisposição Genética para Doença/genética , Transtornos Relacionados ao Uso de Opioides/genética , Limiar da Dor/fisiologia , Dor/tratamento farmacológico , Dor/genética , Animais , Doença Crônica/tratamento farmacológico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/genética , Tolerância a Medicamentos/fisiologia , Adjuvante de Freund/farmacologia , Genótipo , Mediadores da Inflamação/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos MRL lpr , Morfina/farmacologia , Naloxona , Antagonistas de Entorpecentes , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Dor/induzido quimicamente , Limiar da Dor/efeitos dos fármacos , Especificidade da Espécie
18.
Pain ; 115(1-2): 182-90, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15836981

RESUMO

Casein kinase 2 (CK2) is a widely expressed protein kinase. Over the last several years a long list of protein substrates has evolved, many of which have proven or hypothesized roles in nociceptive signal transmission. However, CK2 has not itself been demonstrated to participate in nociception prior to this time. We set out to test the hypothesis that spinal CK2 regulates nociception using several pain models. Our first studies focused on the ability of the selective CK2 inhibitors 4,5,6,7-tetrabromobenzotriazole (TBBT) and 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) to reduce formalin-stimulated pain behaviors in mice. Both phases of the response to subcutaneous formalin were strongly inhibited by intrathecal administration of TBBT or DRB in dose-dependent fashion. Likewise, using the complete Freund's adjuvant (CFA) model of chronic inflammatory pain, TBBT was observed to strongly reduce mechanical allodynia. The inhibition of spinal CK2 with either inhibitor did not, however, alter withdrawal latencies in the hotplate thermal pain model while intrathecal morphine was very effective. Immunohistochemical studies demonstrated all three known CK2 subunits, alpha, alpha' and beta to be expressed in spinal cord tissue as did real-time PCR experiments. While mRNA levels for each of the subunits was transiently enhanced after formalin or CFA hindpaw injection, overall spinal cord protein levels were not elevated in a sustained fashion. Our results indicate that CK2 participates in inflammatory nociception both in the acute and chronic phases. Simple changes in the abundance of spinal CK2 subunits do not likely underlie these phenomena, however.


Assuntos
Caseína Quinase II/metabolismo , Hiperalgesia/metabolismo , Inflamação/metabolismo , Nociceptores/metabolismo , Medula Espinal/metabolismo , Transmissão Sináptica , Animais , Modelos Animais de Doenças , Formaldeído , Hiperalgesia/induzido quimicamente , Hiperalgesia/etiologia , Inflamação/induzido quimicamente , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Limiar da Dor/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Distribuição Tecidual
19.
Mol Med Rep ; 12(1): 1225-32, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25816023

RESUMO

Lumbar disc herniation (LDH) is a term used for a group of conditions, including back pain, femoral nerve pain and sciatica. Currently available treatments and surgical options are insufficient for patients with LDH. Fructus Ligustri Lucidi (FLL) is a herb that is used for treating age-associated diseases. The results of the present study suggested that FLL may be used for treatment of patients with LDH. In the present study, matrix metalloproteinase-1, -3, -8 and -9 (MMP-1, -3, -8 and -9) protein and mRNA expression downregulation was observed in patients with LDH according to western blotting and reverse transcription-quantitative polymerase chain reaction. By contrast, upregulation of interleukin-2 (IL-2), IL-6, IL-8 and tumor necrosis factor-α (TNF-α) expression was observed in patients with LDH, according to an enzyme-linked immunosorbent assay. Mechanical allodynia was observed in rats with LDH not treated with FLL; however, not in FLL­treated rats. IL-2, IL-6, IL-8 and TNF-α expression levels in the serum from untreated rats were significantly higher than that of the FLL­treated rat models. Protein expression levels of MMPs in FLL-treated rats were lower than those in untreated rats. However, the mechanisms underlying the association between FLL and protein expression levels require further investigation.


Assuntos
Hiperalgesia/prevenção & controle , Deslocamento do Disco Intervertebral/tratamento farmacológico , Ligustrum/química , Vértebras Lombares/efeitos dos fármacos , Extratos Vegetais/farmacologia , Adulto , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Ontologia Genética , Humanos , Hiperalgesia/genética , Hiperalgesia/patologia , Hiperalgesia/cirurgia , Interleucina-2/sangue , Interleucina-2/genética , Interleucina-6/sangue , Interleucina-6/genética , Interleucina-8/sangue , Interleucina-8/genética , Deslocamento do Disco Intervertebral/genética , Deslocamento do Disco Intervertebral/patologia , Deslocamento do Disco Intervertebral/cirurgia , Vértebras Lombares/inervação , Vértebras Lombares/patologia , Vértebras Lombares/cirurgia , Masculino , Metaloproteinases da Matriz Secretadas/sangue , Metaloproteinases da Matriz Secretadas/genética , Anotação de Sequência Molecular , Extratos Vegetais/química , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética
20.
Neurosci Lett ; 365(1): 73-7, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15234476

RESUMO

The chronic administration of morphine and related opioid drugs results in tolerance and dependence which reduces the clinical utility of these agents. The CO/NO-cGMP signal transduction cascade plays an important role in morphine tolerance. Principal components of this pathway include heme oxygenase (HO), nitric oxide synthase (NOS), soluble guanylate cyclase (sGC) and cyclic GMP-dependent protein kinase (cGK). We measured and compared the spinal gene expression patterns of these key components using real-time PCR and Western blot analysis after chronic morphine treatment in mice. Our findings indicate that the CO/NO-cGMP signaling pathway is upregulated at multiple points after morphine exposure demonstrating a coordinated molecular and biochemical response. These findings underscore the importance of this signaling pathway in the neuroplastic events occurring during chronic opioid exposure and the value of analyzing the participation of multiple components of a signaling pathway simultaneously rather than individual members in isolation.


Assuntos
Tolerância a Medicamentos/fisiologia , Morfina/farmacologia , Entorpecentes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Monóxido de Carbono/metabolismo , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/biossíntese , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Guanilato Ciclase , Heme Oxigenase (Desciclizante)/biossíntese , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Guanilil Ciclase Solúvel , Medula Espinal/enzimologia , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA