Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
2.
Science ; 276(5312): 586-9, 1997 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-9110976

RESUMO

Spinal cord injuries result in paralysis, because when damaged neurons die they are not replaced. Neurogenesis of electrophysiologically functional neurons occurred in spinal cord cultured from postnatal rats. In these cultures, the numbers of immunocytochemically identified neurons increased over time. Additionally, neurons identified immunocytochemically or electrophysiologically incorporated bromodeoxyuridine, confirming they had differentiated from mitotic cells in vitro. These findings suggest that postnatal spinal cord retains the capacity to generate functional neurons. The presence of neuronal precursor cells in postnatal spinal cord may offer new therapeutic approaches for restoration of function to individuals with spinal cord injuries.


Assuntos
Neurônios/citologia , Medula Espinal/citologia , Potenciais de Ação , Animais , Bromodesoxiuridina/metabolismo , Diferenciação Celular , Células Cultivadas , Meios de Cultura , Proteína Glial Fibrilar Ácida/análise , Imuno-Histoquímica , Mitose , Neurônios/química , Neurônios/metabolismo , Fosfopiruvato Hidratase/análise , Ratos , Medula Espinal/química , Tubulina (Proteína)/análise
3.
Neuron ; 8(2): 211-6, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1371216

RESUMO

Long-term potentiation is a long-lasting, use-dependent increase in the strength of synaptic connections. We investigated the role of nitric oxide (NO) in determining the duration of potentiation induced by high frequency stimulation of afferents in the CA1 region of the rat hippocampus. The calcium/calmodulin-dependent production of NO can be initiated by activation of excitatory amino acid receptors and results in increased levels of cGMP in target cells. Here we report that only a relatively short-term potentiation can be induced in the presence of nitro-L-arginine methyl ester (L-NAME), an NO synthase inhibitor. The effects of L-NAME on the duration of potentiation are partially reversed by coadministration of L-arginine, a precursor of neuronal NO, and by dibutyryl cGMP. Hemoglobin, which binds extracellular NO, also shortens the duration of stimulus-induced potentiation. The results suggest a role for NO in the maintenance of activity-dependent synaptic enhancements, possibly via the generation of cGMP.


Assuntos
Hipocampo/fisiologia , Óxido Nítrico/farmacologia , Aminoácido Oxirredutases/antagonistas & inibidores , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Cálcio/fisiologia , Calmodulina/fisiologia , GMP Cíclico/metabolismo , Dibutiril GMP Cíclico/farmacologia , Hemoglobinas/farmacologia , Hipocampo/metabolismo , Masculino , NG-Nitroarginina Metil Éster , Plasticidade Neuronal/fisiologia , Óxido Nítrico Sintase , Ratos , Sinapses/fisiologia , Fatores de Tempo
4.
J Neurosci ; 21(23): 9355-66, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11717369

RESUMO

This paper describes a model of tumor-induced bone destruction and hyperalgesia produced by implantation of fibrosarcoma cells into the mouse calcaneus bone. Histological examination indicates that tumor cells adhere to the bone edge as early as post-implantation day (PID) 3, but osteolysis does not begin until PID 6, correlating with the development of hyperalgesia. C3H/He mice exhibit a reproducible hyperalgesia to mechanical and cold stimuli between PID 6 and 16. These behaviors are present but significantly reduced with subcutaneous implantation that does not involve bone. Systemic administration of morphine (ED(50) 9.0 mg/kg) dose-dependently attenuated the mechanical hyperalgesia. In contrast, bone destruction and hypersensitivity were not evident in mice implanted with melanoma tumors or a paraffin mass of similar size. A novel microperfusion technique was used to identify elevated levels of the putative algogen endothelin (ET) in perfusates collected from the tumor sites of hyperalgesic mice between PID 7 and 12. Increased ET was evident in microperfusates from fibrosarcoma tumor-implanted mice but not from melanoma tumor-implanted mice, which are not hyperalgesic. Intraplantar injection of ET-1 in naive and, to a greater extent, fibrosarcoma tumor-bearing mice produced spontaneous pain behaviors, suggesting that ET-1 activates primary afferent fibers. Intraplantar but not systemic injection of the ET-A receptor antagonist BQ-123 partially blocked tumor-associated mechanical hyperalgesia, indicating that ET-1 contributes to tumor-induced nociception. This model provides a unique approach for quantifying the behavioral, biochemical, and electrophysiological consequences of tumor-nerve interactions.


Assuntos
Modelos Animais de Doenças , Fibrossarcoma/fisiopatologia , Melanoma Experimental/fisiopatologia , Neoplasias Experimentais/fisiopatologia , Dor/fisiopatologia , Nervos Periféricos/fisiopatologia , Animais , Comportamento Animal , Calcâneo/patologia , Calcâneo/cirurgia , Cruzamentos Genéticos , Endotelina-1/efeitos adversos , Endotelina-1/biossíntese , Endotelina-1/metabolismo , Fibrossarcoma/complicações , Fibrossarcoma/patologia , Membro Posterior/patologia , Membro Posterior/fisiopatologia , Hiperalgesia/diagnóstico , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Melanoma Experimental/complicações , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Experimentais/complicações , Neoplasias Experimentais/patologia , Dor/diagnóstico , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Nervos Periféricos/patologia , Estimulação Física , Células Tumorais Cultivadas
5.
J Neurosci ; 21(23): 9367-76, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11717370

RESUMO

We used a murine model to investigate functional interactions between tumors and peripheral nerves that may contribute to pain associated with cancer. Implantation of fibrosarcoma cells in and around the calcaneus bone produced mechanical hyperalgesia of the ipsilateral paw. Electrophysiological recordings from primary afferent fibers in control and hyperalgesic mice with tumor revealed the development of spontaneous activity (0.2-3.4 Hz) in 34% of cutaneous C-fibers adjacent to the tumor (9-17 d after implantation). C-fibers in tumor-bearing mice exhibited a mean decrease in heat threshold of 3.5 +/- 0.10 degrees C. We also examined innervation of the skin overlying the tumor. Epidermal nerve fibers (ENFs) were immunostained for protein gene product 9.5, imaged using confocal microscopy, and analyzed in terms of number of fibers per millimeter of epidermal length and branching (number of nodes per fiber). Divergent morphological changes were linked to tumor progression. Although branching of ENFs increased significantly relative to control values, in later stages (16-24 d after implantation) of tumor growth a sharp decrease in the number of ENFs was observed. This decay of epidermal innervation of skin over the tumor coincided temporally with gradual loss of electrophysiological activity in tumor-bearing mice. The development of spontaneous activity and sensitization to heat in C-fibers and increased innervation of cutaneous structures within the first 2 weeks of tumor growth suggest activation and sensitization of a proportion of C-fibers. The decrease in the number of ENFs observed in later stages of tumor development implicates neuropathic involvement in this model of cancer pain.


Assuntos
Modelos Animais de Doenças , Fibrossarcoma/fisiopatologia , Neoplasias Experimentais/fisiopatologia , Fibras Nervosas , Neurônios Aferentes , Dor/fisiopatologia , Nervos Periféricos/fisiopatologia , Animais , Calcâneo/patologia , Calcâneo/cirurgia , Progressão da Doença , Eletrofisiologia , Epiderme/inervação , Epiderme/patologia , Epiderme/fisiopatologia , Fibrossarcoma/complicações , Fibrossarcoma/patologia , Membro Posterior/patologia , Membro Posterior/fisiopatologia , Hiperalgesia/diagnóstico , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Experimentais/complicações , Neoplasias Experimentais/patologia , Fibras Nervosas/patologia , Neurônios Aferentes/patologia , Dor/diagnóstico , Dor/etiologia , Medição da Dor , Nervos Periféricos/patologia , Estimulação Física , Células Tumorais Cultivadas
6.
Neuroscience ; 132(2): 479-91, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15802198

RESUMO

A nociceptive role for tumor necrosis factor-alpha (TNF-alpha) in naive mice and in mice with fibrosarcoma tumor-induced primary hyperalgesia was investigated. The presence of TNF-alpha mRNA was confirmed in tumor site homogenates by reverse transcription-polymerase chain reaction (RT-PCR), and examination of TNF-alpha protein levels in tumor-bearing mice indicated a significantly higher concentration of this cytokine in tumor microperfusates and tumor site homogenates compared with that obtained from a similar site on the contralateral limb or in naive mice. Intraplantar injection of TNF-alpha into naive or fibrosarcoma tumor-bearing mice induced mechanical hypersensitivity, as measured by withdrawal responses evoked by von Frey monofilaments. This hypersensitivity suggests that TNF-alpha can excite or sensitize primary afferent fibers to mechanical stimulation in both naive and tumor-bearing mice. In addition, the hyperalgesia produced by TNF-alpha was completely eliminated when the injected TNF-alpha was pre-incubated with the soluble receptor antagonist TNFR:Fc. Importantly, pre-implantation systemic as well as post-implantation intra-tumor injection of TNFR:Fc partially blocked the mechanical hyperalgesia, indicating that local production of TNF-alpha may contribute to tumor-induced nociception.


Assuntos
Fibrossarcoma/metabolismo , Neoplasias Experimentais/metabolismo , Dor/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Comportamento Animal , Linhagem Celular Tumoral , Fibrossarcoma/complicações , Regulação Neoplásica da Expressão Gênica/fisiologia , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Experimentais/complicações , Dor/etiologia , Medição da Dor/métodos , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Necrose Tumoral alfa/genética
7.
Br J Pharmacol ; 172(2): 388-402, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24641506

RESUMO

UNLABELLED: Opioid and α2 -adrenoceptor agonists are potent analgesic drugs and their analgesic effects can synergize when co-administered. These supra-additive interactions are potentially beneficial clinically; by increasing efficacy and/or reducing the total drug required to produce sufficient pain relief, undesired side effects can be minimized. However, combination therapies of opioids and α2 -adrenoceptor agonists remain underutilized clinically, in spite of a large body of preclinical evidence describing their synergistic interaction. One possible obstacle to the translation of preclinical findings to clinical applications is a lack of understanding of the mechanisms underlying the synergistic interactions between these two drug classes. In this review, we provide a detailed overview of the interactions between different opioid and α2 -adrenoceptor agonist combinations in preclinical studies. These studies have identified the spinal cord as an important site of action of synergistic interactions, provided insights into which receptors mediate these interactions and explored downstream signalling events enabling synergy. It is now well documented that the activation of both µ and δ opioid receptors can produce synergy with α2 -adrenoceptor agonists and that α2 -adrenoceptor agonists can mediate synergy through either the α2A or the α2C adrenoceptor subtypes. Current hypotheses surrounding the cellular mechanisms mediating opioid-adrenoceptor synergy, including PKC signalling and receptor oligomerization, and the evidence supporting them are presented. Finally, the implications of these findings for clinical applications and drug discovery are discussed. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Assuntos
Receptores Adrenérgicos alfa 2/metabolismo , Receptores Opioides/metabolismo , Agonistas alfa-Adrenérgicos/farmacocinética , Agonistas alfa-Adrenérgicos/farmacologia , Analgesia , Analgésicos Opioides/farmacologia , Animais , Sinergismo Farmacológico , Humanos
8.
Br J Pharmacol ; 172(2): 642-53, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24827408

RESUMO

BACKGROUND AND PURPOSE: We recently found that PKCε was required for spinal analgesic synergy between two GPCRs, δ opioid receptors and α2 A adrenoceptors, co-located in the same cellular subpopulation. We sought to determine if co-delivery of µ and δ opioid receptor agonists would similarly result in synergy requiring PKCε. EXPERIMENTAL APPROACH: Combinations of µ and δ opioid receptor agonists were co-administered intrathecally by direct lumbar puncture to PKCε-wild-type (PKCε-WT) and -knockout (PKCε-KO) mice. Antinociception was assessed using the hot-water tail-flick assay. Drug interactions were evaluated by isobolographic analysis. KEY RESULTS: All agonists produced comparable antinociception in both PKCε-WT and PKCε-KO mice. Of 19 agonist combinations that produced analgesic synergy, only 3 required PKCε for a synergistic interaction. In these three combinations, one of the agonists was morphine, although not all combinations involving morphine required PKCε. Morphine + deltorphin II and morphine + deltorphin I required PKCε for synergy, whereas a similar combination, morphine + deltorphin, did not. Additionally, morphine + oxymorphindole required PKCε for synergy, whereas a similar combination, morphine + oxycodindole, did not. CONCLUSIONS AND IMPLICATIONS: We discovered biased agonism for a specific signalling pathway at the level of spinally co-delivered opioid agonists. As the bias is only revealed by an appropriate ligand combination and cannot be accounted for by a single drug, it is likely that the receptors these agonists act on are interacting with each other. Our results support the existence of µ and δ opioid receptor heteromers at the spinal level in vivo. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Dor/tratamento farmacológico , Proteína Quinase C-épsilon/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Temperatura Alta , Ligantes , Masculino , Camundongos Knockout , Morfina/farmacologia , Morfina/uso terapêutico , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Dor/metabolismo , Multimerização Proteica , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Medula Espinal/metabolismo
9.
Pain ; 66(2-3): 331-41, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8880857

RESUMO

Formalin injected subcutaneously into the hindpaw of the rat produces an animal model of inflammation that exhibits a phasic component and a tonic component of pain. We evaluated the effects of a nitric oxide synthase inhibitor, N omega-nitro-L-arginine methyl ester (L-NAME), on a formalin-induced behavior, hindpaw licking, and on Fos-labeling of nuclei in the fifth lumbar spinal segment. Our results demonstrated that pretreatment with intrathecal doses of 0.3 and 1.0 mg of L-NAME significantly reduced licking behavior associated with injection of formalin into the left hindpaw of the rat. In addition, these same doses of L-NAME reduced formalin-induced Fos-labeling in the ipsilateral dorsal gray matter (as compared to the contralateral gray matter). Qualitative assessment suggested that the reduction in labeling occurred primarily in the superficial dorsal horn. The stereoisomer, D-NAME, administered at the same doses had little to no effect on either formalin-induced licking or Fos-labeling. Finally, our results revealed that total licking time was related to Fos-labeling. Rats that spent less time licking the hindpaw exhibited a smaller increase in Fos-labeling. Our results suggest that the production of nitric oxide is associated with licking behavior resulting from formalin injection into the hindpaw of rats. Our results also suggest that the production of nitric oxide and Fos are associated. Indeed, these substances may be involved in spinal pathways associated with nociception.


Assuntos
Comportamento Animal/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inflamação/metabolismo , Inflamação/psicologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fos/biossíntese , Medula Espinal/metabolismo , Sequência de Aminoácidos , Animais , Inibidores Enzimáticos/administração & dosagem , Formaldeído , Hiperalgesia/metabolismo , Hiperalgesia/psicologia , Imuno-Histoquímica , Inflamação/induzido quimicamente , Masculino , Dados de Sequência Molecular , NG-Nitroarginina Metil Éster/administração & dosagem , Medição da Dor/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos
10.
Pain ; 80(1-2): 37-43, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10204716

RESUMO

The redox modulatory site of the N-methyl-D-aspartate (NMDA) receptor directly regulates NMDA receptor function. Sulfhydryl reducing agents, such as dithiothreitol (DTT), potentiate NMDA receptor-evoked currents in vitro, whereas oxidizing agents, such as 5,5'-dithio-bis-(2-nitrobenzoic acid) (DTNB), attenuate these currents. In this study, we examined the effect of this redox manipulations on nociceptive spinal cord signaling in mice. Intrathecal (i.t.) administration of DTT (0.1-30 nmol), presumably reducing the NMDA receptor, dose-dependently enhanced NMDA-induced nociceptive behaviors, and this enhancement was blocked by the oxidizing agent, DTNB. Pretreatment with DTT (10 nmol, i.t.) enhanced NMDA-induced tail-flick thermal hyperalgesia and intraplantar formalin-induced nociceptive behaviors. Finally, DTT pretreatment enhanced the long lasting allodynia induced by i.t. administration of dynorphin, whereas post-treatment with DTNB reduced the permanent allodynia induced by dynorphin for 5 days. Potentiation of all four of these NMDA-dependent nociceptive behaviors by DTT suggests that the reduction of the NMDA receptor by endogenous reducing agents may contribute to augmented pain transmission in response to activation by endogenous glutamate. Moreover, blockade of in vivo NMDA receptor reducing agents or oxidation of the NMDA receptor redox site may prove therapeutically useful in the treatment of chronic pain.


Assuntos
Dinorfinas , Dor/fisiopatologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Doença Aguda , Animais , Ácido Ditionitrobenzoico/administração & dosagem , Ácido Ditionitrobenzoico/farmacologia , Ditiotreitol/administração & dosagem , Ditiotreitol/farmacologia , Dinorfinas/administração & dosagem , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Agonistas de Aminoácidos Excitatórios/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos ICR , N-Metilaspartato/administração & dosagem , N-Metilaspartato/farmacologia , Oxirredução , Dor/induzido quimicamente , Medição da Dor , Receptores de N-Metil-D-Aspartato/metabolismo , Substâncias Redutoras/administração & dosagem , Substâncias Redutoras/farmacologia , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiopatologia , Reagentes de Sulfidrila/administração & dosagem , Reagentes de Sulfidrila/farmacologia
11.
Pain ; 84(2-3): 159-67, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10666520

RESUMO

Dynorphin A is an endogenous opioid peptide, which has previously been shown to produce a long-lasting allodynia and hyperalgesia in mice, behavioral states consistent with signs of clinically observed neuropathic pain. This dynorphin-induced allodynia was used as a pharmacological, central model of neuropathic pain. In this study, we examined the involvement of the cytokine IL-1beta, the transcription factor nuclear factor kappa B (NF-kappaB), and de novo protein synthesis in the development of allodynia induced by intrathecal (i.t.) administration of dynorphin in male ICR mice. Pretreatment with the protein synthesis inhibitor cycloheximide (0. 3-85nmol), the NF-kappaB inhibitor pyrrolidinedithiocarbamate (PDTC) (0.001-1000pmol), the IL-1 receptor antagonist (IL-1ra) protein (0. 01-100ng), the caspase-1 inhibitor (YVAD) (0.1-300pmol), and the anti-inflammatory cytokine IL-10 (0.1-300ng) all dose-dependently reduced the induction of dynorphin-induced allodynia. Finally, IL-10 administered within the first 24h after the dynorphin insult prevented the development of chronic allodynia. These results demonstrate that the anti-inflammatory cytokines IL-10 and IL-1ra impede the development of dynorphin-induced allodynia. These results also suggest that production of new proteins through NF-kappaB activation is required for the induction of allodynia. We speculate that IL-1ra, IL-10, PDTC and cycloheximide interfere with the central pro-inflammatory cascade. Modulation of cytokine activity in the spinal cord may therefore prove to be an effective therapeutic strategy for the treatment of chronic pain.


Assuntos
Citocinas/fisiologia , Dinorfinas , Hiperestesia/induzido quimicamente , Hiperestesia/fisiopatologia , Animais , Interleucina-1/fisiologia , Interleucina-10/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/fisiologia , Biossíntese de Proteínas
12.
Pain ; 72(1-2): 253-60, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9272810

RESUMO

The endogenous opioid peptide dynorphin A has non-opioid effects that can damage the spinal cord when given in high doses. Dynorphin has been shown to increase the receptive field size of spinal cord neurons and facilitate C-fiber-evoked reflexes. Furthermore, endogenous dynorphin levels increase following damage to the spinal cord, injury to peripheral nerves, or inflammation. In this study, sensory processing was characterized following a single, intrathecal injection of dynorphin A (1-17) in mice. A single intrathecal injection of dynorphin A (1-17) (3 nmol, i.t.) induced mechanical allodynia (hind paw, von Frey filaments) lasting 70 days, tactile allodynia (paint brush applied to flank) lasting 14 days, and cold allodynia (acetone applied to the dorsal hind paw) lasting 7 days. Similarly, dynorphin A (2-17) (3 nmol, i.t.), a non-opioid peptide, induced cold and tactile allodynia analogous to that induced by dynorphin A (1-17), indicating the importance of non-opioid receptors. Pretreatment with the NMDA antagonists, MK-801 and LY235959, but not the opioid antagonist, naloxone, blocked the induction of allodynia. Post-treatment with MK-801 only transiently blocked the dynorphin-induced allodynia, suggesting the NMDA receptors may be involved in the maintenance of allodynia as well as its induction. We have induced a long-lasting state of allodynia and hyperalgesia by a single intrathecal injection of dynorphin A (1-17) in mice. The allodynia induced by dynorphin required NMDA receptors rather than opioid receptors. This result is consistent with results in rats and with signs of clinically observed neuropathic pain. This effect of exogenously administered dynorphin raises the possibility that increased levels of endogenous dynorphins associated with spinal cord injuries may participate in the genesis and maintenance of neuropathic pain.


Assuntos
Dinorfinas/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Dor/induzido quimicamente , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores Opioides/fisiologia , Animais , Maleato de Dizocilpina/farmacologia , Injeções Espinhais , Isoquinolinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Antagonistas de Entorpecentes , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
13.
Pain ; 68(2-3): 275-81, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9121815

RESUMO

Neuropathic pain states are accompanied by increased sensitivity to both noxious and non-noxious sensory stimuli, characterized as hyperalgesia and allodynia, respectively. In animal models of neuropathic pain, the presence of hyperalgesia and allodynia are accompanied by neuroplastic changes including increased spinal levels of substance P, cholecystokinin (CCK), and dynorphin. N-Methyl-D-aspartate (NMDA) receptors appear to be involved in maintaining the central sensitivity which contributes to neuropathic pain. In addition to its opioid activities, dynorphin has been suggested to act at the NMDA receptor complex. In an attempt to mimic the increased levels of spinal dynorphin seen in animal models of neuropathic pain, rats received a single intrathecal (i.t.) injection of dynorphin A(1-17), dynorphin A(1-13), dynorphin A(2-17) or dynorphin A(2-13) through indwelling catheters. Tactile allodynia was determined by measuring response threshold to probing with von Frey filaments. Dynorphin A(1-17) administration evoked significant and long-lasting tactile allodynia (i.e. > 60 days). Likewise, the i.t. administration of dynorphin A(1-13) or dynorphin A(2-17) or dynorphin A(2-13) also produced long-lasting tactile allodynia. Intrathecal pretreatment, but not post-treatment, with MK-801 prevented dynorphin A(1-17)-induced development of allodynia; i.t. administration of MK-801 alone had no effect on responses to tactile stimuli. In contrast, i.t. pretreatment with naloxone did not affect the development of tactile allodynia induced by dynorphin A(1-17) or alter sensory threshold when given alone. These results demonstrate that a single dose of dynorphin A, or its des-Tyr fragments, produces long-lasting allodynia which may be irreversible in the rat. Further, this effect appears to be mediated through activation of NMDA, rather than opioid, receptors. While the precise mechanisms underlying the development and maintenance of the allodynia is unclear, it seems possible that dynorphin may produce changes in the spinal cord, which may contribute to the development of signs reminiscent of a "neuropathic' state. Given that levels of dynorphin are elevated following nerve injury, it seems reasonable to speculate that dynorphin may have a pathologically relevant role in neuropathic pain states.


Assuntos
Dinorfinas/antagonistas & inibidores , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Fragmentos de Peptídeos/antagonistas & inibidores , Limiar Sensorial/efeitos dos fármacos , Animais , Doença Crônica , Maleato de Dizocilpina/uso terapêutico , Injeções Espinhais , Masculino , Naloxona/uso terapêutico , Dor/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Ratos , Ratos Sprague-Dawley
14.
Neuropharmacology ; 21(8): 745-50, 1982 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7121746

RESUMO

The antinociceptive effectiveness of spinally administered opiates raises the question of their neural target in the spinal cord. A large body of evidence points to a site of action on primary afferent fibers before the first central synapse. The dorsal root potential (DRP) is a consequence of phasic presynaptic inhibitory action on the primary afferent fibers. In these studies in the rat, dorsal root potentials, evoked by non-noxious subcutaneous electrical stimulation, were altered by systemic and spinal administration of opiates and by two types of noxious stimulation. Noxious stimulation was accompanied by increased dorsal root potential amplitude, whereas antinociception was associated with decreased dorsal root potential amplitude. Treatment with morphine decreased the dorsal potential and blocked the increase in amplitude induced by noxious by noxious stimuli. These results indicate that nociception and antinociception differentially modulate the phasic component of presynaptic inhibition on primary afferent fibers in the spinal cord.


Assuntos
Analgésicos/farmacologia , Nociceptores/fisiologia , Raízes Nervosas Espinhais/fisiologia , Animais , Bradicinina/farmacologia , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Masculino , Morfina/farmacologia , Naloxona/farmacologia , Entorpecentes/farmacologia , Ratos , Ratos Endogâmicos , Raízes Nervosas Espinhais/efeitos dos fármacos
15.
Neuropharmacology ; 23(12A): 1415-8, 1984 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6335574

RESUMO

The endogenous compounds, serotonin (5-HT) and tryptamine (TA), have been found to alter function in the CNS, supporting the hypothesis that they serve as neurotransmitters. It is reported here that intrathecal injections of either 5-HT or tryptamine elicited similar behavioral syndromes consisting of caudally-directed biting, or licking and scratching. Serotonin was found to be considerably more potent than tryptamine. However, administration of both indoleamines produced synergistic interactions with respect to this behavioral syndrome. Administration of fluoxetine with either 5-HT or tryptamine potentiated the scratching behavior produced by either indoleamine, but failed to enhance the effect of an injection of 5-HT plus tryptamine. Administration of methysergide blocked the effects of both indolamines. The results are discussed in terms of a possible mechanism of the synergism between 5-HT and tryptamine.


Assuntos
Comportamento Animal/efeitos dos fármacos , Serotonina/farmacologia , Triptaminas/farmacologia , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Fluoxetina/farmacologia , Injeções Espinhais , Masculino , Metisergida/farmacologia , Camundongos
16.
Neuroscience ; 19(2): 393-401, 1986 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3774148

RESUMO

The importance of the spinothalamic tract in pain transmission makes it an attractive candidate for study with respect to the effects of antinociceptive compounds. We have been interested in the analgesic actions of opioids and noradrenergic agents at the spinal level and have investigated the effects of these agents on extracellularly recorded nociceptive dorsal horn neurons in the rat. Spinothalamic tract cells were identified by antidromic activation from the somatosensory thalamus. Morphine was administered by bathing the spinal cord in an artificial cerebrospinal fluid solution which contained a known concentration of drug. We observed a dose-related inhibition, naloxone-reversible in some cases, of activity produced by spinally administered morphine in identified rat spinothalamic tract cells and dorsal horn nociceptive neurons. Morphine had no effect on stimulus-evoked responses of low threshold dorsal horn neurons.


Assuntos
Morfina/farmacologia , Nociceptores/efeitos dos fármacos , Dor/prevenção & controle , Medula Espinal/efeitos dos fármacos , Tratos Espinotalâmicos/efeitos dos fármacos , Administração Tópica , Animais , Injeções Intravenosas , Masculino , Morfina/administração & dosagem , Ratos , Ratos Endogâmicos
17.
Neuroscience ; 93(4): 1399-407, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10501465

RESUMO

Neuropathic pain resulting from peripheral nerve injury can often be relieved by administration of alpha-adrenergic receptor antagonists. Tonic activation of alpha-adrenergic receptors may therefore facilitate the hyperalgesia and allodynia associated with neuropathic pain. It is currently unclear whether alpha2A- or alpha2c-adrenergic receptor subtypes are involved in the pro-nociceptive actions of alpha-adrenergic receptors under neuropathic conditions. We therefore investigated the effects of peripheral nerve injury on the expression of these subtypes in rat spinal cord using immunohistochemical techniques. In addition, neuropeptide Y immunoreactivity was examined as an internal control because it has previously been shown to be up-regulated following nerve injury. We observed a decrease in alpha2A-adrenergic receptor immunoreactivity in the spinal cord ipsilateral to three models of neuropathic pain: complete sciatic nerve transection, chronic constriction injury of the sciatic nerve and L5/L6 spinal nerve ligation. The extent of this down-regulation was significantly correlated with the magnitude of injury-induced changes in mechanical sensitivity. In contrast, alpha2c-adrenergic receptor immunoreactivity was only increased in the spinal nerve ligation model; these increases did not correlate with changes in mechanical sensitivity. Neuropeptide Y immunoreactivity was up-regulated in all models examined. Increased expression of neuropeptide Y correlated with changes in mechanical sensitivity. The decrease in alpha2A-adrenergic receptor immunoreactivity and the lack of consistent changes in alpha2C-adrenergic receptor immunoreactivity suggest that neither of these receptor subtypes is likely to be responsible for the abnormal adrenergic sensitivity observed following nerve injury. On the contrary, the decrease in alpha2A-adrenergic receptor immunoreactivity following nerve injury may result in an attenuation of the influence of descending inhibitory noradrenergic input into the spinal cord resulting in increased excitatory transmitter release following peripheral stimuli.


Assuntos
Receptores Adrenérgicos alfa 2/análise , Nervo Isquiático/lesões , Medula Espinal/química , Nervos Espinhais/lesões , Animais , Doença Crônica , Hiperalgesia/fisiopatologia , Imuno-Histoquímica , Ligadura , Masculino , Síndromes de Compressão Nervosa/fisiopatologia , Neuropeptídeo Y/análise , Dor/fisiopatologia , Estimulação Física , Ratos , Ratos Sprague-Dawley
18.
Brain Res Mol Brain Res ; 28(1): 72-80, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7707880

RESUMO

We functionally expressed alpha 2-adrenergic, beta 2-adrenergic, and delta-opioid receptors in Xenopus laevis oocytes. We detected receptor function as changes in currents carried by adenosine 3',5'-cyclic monophosphate (cAMP)-regulated chloride channels provided by the cystic fibrosis transmembrane conductance regulator (CFTR) and recorded by two-electrode voltage clamp. Co-application of forskolin and isobutylmethylxanthine (IBMX) or IBMX alone produced currents with a reversal potential indicative of chloride ions only in oocytes previously injected with mRNA encoding CFTR. Isoproterenol produced concentration-dependent responses in oocytes injected with mRNA encoding beta 2-adrenergic receptors and CFTR, and co-administration of propranolol antagonized these responses. Similarly, the alpha 2-adrenergic agonist UK14304 increased IBMX-induced currents only in oocytes injected with mRNA encoding alpha 2-adrenergic receptors and CFTR, and idazoxan antagonized these enhancements. The delta-opioid agonist DADLE produced concentration-related, naloxone-reversible increases in IBMX- and forskolin-induced currents only in oocytes injected with mRNA encoding delta-opioid receptors and CFTR. In oocytes co-injected with alpha 2, beta 2, and CFTR mRNAs, isobolographic analysis revealed an additive interaction between alpha 2- and beta 2-adrenergic receptors. These studies establish the oocyte as a cell system for studying the interactions among cAMP-modulating G protein-coupled receptors and provide another example of alternative coupling of alpha 2-adrenergic and delta-opioid receptors to G proteins, possibly Gs proteins, other than Gi proteins.


Assuntos
Isoproterenol/farmacologia , Naloxona/farmacologia , Propranolol/farmacologia , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Adenilil Ciclases , Animais , Tartarato de Brimonidina , AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico , Fibrose Cística , Relação Dose-Resposta a Droga , Modelos Biológicos , Oócitos , Quinoxalinas/farmacologia , RNA Mensageiro , Receptores Opioides , Xenopus laevis
19.
Brain Res Mol Brain Res ; 44(1): 55-65, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9030698

RESUMO

The objective of this study was to characterize the signaling mechanisms of the mu-opioid receptor in its coupling to the cystic fibrosis transmembrane conductance regulator (CFTR) when coexpressed in Xenopus oocytes. Because oocytes do not contain endogenous cAMP-regulated ion channels, the cAMP-modulated CFTR was coexpressed with receptors as a 'reporter' channel. Agonist treatment of oocytes coexpressing mu-opioid receptors, beta2-adrenergic receptors and CFTR produced Cl- currents in a dose-related manner and immunocytochemical analysis confirmed receptor expression. These data suggest that opioid agonists could activate adenylyl cyclase in this system to elevate cAMP levels. Heterotrimeric G protein betagamma-subunits acting on adenylyl cyclase type II would increase cAMP levels. The probable presence of adenylyl cyclase type II and other components of opioid signal transduction such as G(i alpha2), were demonstrated by RT-PCR. However, measurement of cAMP levels in individual oocytes by radioimmunoassay showed that opioid agonist application to oocytes expressing mu-opioid receptors, beta2-adrenergic receptors and CFTR did not increase cAMP levels, whereas application of the beta2-adrenergic agonist, isoproterenol, or IBMX alone did increase cAMP levels. Opioid-induced CFTR activation was not affected by either application of the broad spectrum kinase inhibitor, H7, nor by application of the specific PKA inhibitor, KT5720. Injection of free betagamma-subunits, which could activate the endogenous type II cyclase, was unable to produce measurable currents in oocytes expressing the CFTR. These studies indicate that opioid activation of the CFTR is not mediated through a cAMP/PKA pathway, by either betagamma-subunit activation of an adenylyl cyclase type II or promiscuous coupling to G(s alpha).


Assuntos
AMP Cíclico/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Sequência de Aminoácidos , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Imuno-Histoquímica , Isoproterenol/farmacologia , Dados de Sequência Molecular , Oócitos/metabolismo , Xenopus
20.
Ann N Y Acad Sci ; 525: 228-36, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-2455461

RESUMO

Compounds that produce depolarization of nociceptive neurons in the dorsal horn of the spinal cord also elicit a rather specific kind of caudally directed biting, licking, and/or scratching behavior when they are injected intrathecally in mice. We sought to use this elicited grooming behavior as a test for compounds that might inhibit the neurons excited by the excitatory agents. All three neurokinins--substance P, neurokinin A (substance K), neurokinin B (neuromedin K)--and excitatory amino acids active at N-methyl-D-aspartate (NMDA) or quisqualate receptors produce similar behaviors, which last for 1 minute after i.t. injection. Our data indicate that mu opioid agonists or alpha adrenergic agonists block both neurokinin-elicited behavior and EAA-elicited behavior; delta opioid agonists block only neurokinin-elicited behavior; and PCP/sigma "opioid" agonists block only EAA-elicited behavior. Somatostatin and serotonin produce qualitatively different behaviors by themselves and, when administered with neurokinins, partially block neurokinin-elicited behavior.


Assuntos
Aminoácidos/farmacologia , Comportamento Animal/efeitos dos fármacos , Asseio Animal/efeitos dos fármacos , Medula Espinal/fisiologia , Substância P/farmacologia , Aminoácidos/antagonistas & inibidores , Animais , Injeções Espinhais , Neurocinina A , Neurocinina B , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/farmacologia , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia , Somatostatina/farmacologia , Substância P/antagonistas & inibidores , Triptaminas/antagonistas & inibidores , Triptaminas/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa