Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Biochem Biophys Res Commun ; 501(2): 514-519, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29738766

RESUMO

Nocistatin (NST) is a neuropeptide produced from the same precursor protein of opioid peptide nociceptin/orphanin FQ, and it is involved in a broad range of central functions including pain transmission in the nervous system. However, the composition and structure of the receptor(s) for NST remain unclear. Here, we developed NST photoaffinity probe to identify NST receptor. The NST photoaffinity probe contains an azide moiety for the tagging of the binding protein as well as biotin for protein detection. Intrathecal administration of a NST photoaffinity probe, biotin-(AC5)2-[Y6,azF14]bNST, inhibited the nociceptin/orphanin FQ-evoked tactile pain allodynia in a manner similar to that of NST. The biotin-(AC5)2-[Y6,azF14]bNST-binding proteins were primarily localized in the gray matter of the spinal cord. After photo-crosslinking of the protein complex with biotin-(AC5)2-[Y6,azF14]bNST, two dominant binding protein bands were observed at 58 and 64 kDa. Thus, biotin-(AC5)2-[Y6,azF14]bNST has pharmacological activity and is useful for characterizing the NST receptor.


Assuntos
Peptídeos Opioides/análise , Marcadores de Fotoafinidade/química , Medula Espinal/química , Animais , Biotinilação , Hiperalgesia/metabolismo , Masculino , Camundongos , Peptídeos Opioides/metabolismo , Marcadores de Fotoafinidade/metabolismo , Ligação Proteica , Medula Espinal/metabolismo
2.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27030720

RESUMO

BACKGROUND: Chronic pain associated with inflammation is an important clinical problem, and the underlying mechanisms remain poorly understood. 4-Nitrophenylphosphatase domain and nonneuronal SNAP25-like protein homolog (NIPSNAP) 1, an interacting protein with neuropeptide nocistatin, is implicated in the inhibition of tactile pain allodynia. Although nocistatin inhibits some inflammatory pain responses, whether NIPSNAP1 affects inflammatory pain appears to be unclear. Here, we examined the nociceptive behavioral response of NIPSNAP1-deficient mice and the expression of NIPSNAP1 following peripheral inflammation to determine the contribution of NIPSNAP1 to inflammatory pain. RESULTS: Nociceptive behavioral response increased in phase II of the formalin test, particularly during the later stage (26-50 min) in NIPSNAP1-deficient mice, although the response during phase I (0-15 min) was not significantly different between the deficient and wild-type mice. Moreover, phosphorylation of extracellular signal-related kinase was enhanced in the spinal dorsal horn of the deficient mice. The prolonged inflammatory pain induced by carrageenan and complete Freund's adjuvant was exacerbated in NIPSNAP1-deficient mice. NIPSNAP1 mRNA was expressed in small- and medium-sized neurons of the dorsal root ganglion and motor neurons of the spinal cord. In the formalin test, NIPSNAP1 mRNA was slightly increased in dorsal root ganglion but not in the spinal cord. In contrast, NIPSNAP1 mRNA levels in dorsal root ganglion were significantly decreased during 24-48 h after carrageenan injection. Prostaglandin E2, a major mediator of inflammation, stimulated NIPSNAP1 mRNA expression via the cAMP-protein kinase A signaling pathway in isolated dorsal root ganglion cells. CONCLUSIONS: These results suggest that changes in NIPSNAP1 expression may contribute to the pathogenesis of inflammatory pain.


Assuntos
Inflamação/complicações , Inflamação/metabolismo , Neuropeptídeos/metabolismo , Peptídeos Opioides/metabolismo , Dor/complicações , Dor/metabolismo , Proteínas/metabolismo , Animais , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/farmacologia , Formaldeído , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Inflamação/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana , Camundongos , Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Eur J Neurosci ; 43(11): 1499-508, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27038234

RESUMO

Pathological chronic pain, which is frequently associated with prolonged tissue damage, inflammation, tumour invasion, and neurodegenerative diseases, gives rise to hyperalgesia and allodynia. We previously reported that intrathecal administration of nociceptin/orphanin FQ (N/OFQ), an endogenous ligand for the orphan opioid receptor-like receptor, in the femtomole range induces touch-evoked allodynia. N/OFQ has been implicated in multiple signalling pathways, such as inhibition of cAMP production and Ca(2+) channels, or activation of K(+) channels and mitogen-activated protein kinase, although the signalling pathways of N/OFQ-induced allodynia remain unclear. To address these issues, we developed an ex vivo mitogen-activated protein kinase assay by using intact slices of mouse spinal cord. N/OFQ markedly increased the phosphorylation of c-Jun N-terminal kinase (JNK) in the superficial dorsal horn of the spinal cord. The N/OFQ-stimulated JNK phosphorylation was significantly inhibited by pertussis toxin, the phospholipase C inhibitor U73122, and the inositol trisphosphate receptor antagonist Xestospongin C. Intrathecal administration of the JNK inhibitor SP600125 inhibited N/OFQ-evoked allodynia. The N/OFQ-induced increase in JNK phosphorylation was observed in astrocytes that expressed glial fibrillary acidic protein. N/OFQ also induced monocyte chemoattractant protein-1 (MCP-1) release via the JNK pathway, and N/OFQ-induced JNK phosphorylation was observed in MCP-1-immunoreactive astrocytes. Intrathecal administration of the MCP-1 receptor antagonist RS504393 inhibited N/OFQ-evoked allodynia. These results suggest that, in the spinal dorsal horn, N/OFQ induces allodynia through activation of JNK via the phospholipase C-inositol trisphosphate pathway, which is coupled to pertussis toxin-sensitive G-protein, and following the release of MCP-1 from astrocytes.


Assuntos
Quimiocina CCL2/metabolismo , Hiperalgesia/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Peptídeos Opioides/administração & dosagem , Medula Espinal/metabolismo , Animais , Astrócitos/metabolismo , Hiperalgesia/induzido quimicamente , Injeções Espinhais , Camundongos , Fosforilação , Transdução de Sinais , Nociceptina
4.
J Biol Chem ; 287(13): 10403-10413, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22311985

RESUMO

4-Nitrophenylphosphatase domain and non-neuronal SNAP25-like protein homolog 1 (NIPSNAP1) is a molecule of physiologically unknown function, although it is predominantly expressed in the brain, spinal cord, liver, and kidney. We identified NIPSNAP1 as a protein that interacts with the neuropeptide nocistatin (NST) from synaptosomal membranes of mouse spinal cord using high-performance affinity latex beads. NST, which is produced from the same precursor protein as an opioid-like neuropeptide nociceptin/orphanin FQ (N/OFQ), has opposite effects on pain transmission evoked by N/OFQ. The calculated full-length pre-protein of NIPSNAP1 was 33 kDa, whereas the N-terminal truncated form of NIPSNAP1 (29 kDa) was ubiquitously expressed in the neuronal tissues, especially in synaptic membrane and mitochondria of brain. The 29-kDa NIPSNAP1 was distributed on the cell surface, and NST interacted with the 29-kDa but not the 33-kDa NIPSNAP1. Although intrathecal injection of N/OFQ induced tactile allodynia in both wild-type and NIPSNAP1-deficient mice, the inhibition of N/OFQ-evoked tactile allodynia by NST seen in wild-type mice was completely lacking in the deficient mice. These results suggest that NIPSNAP1 is an interacting molecule of NST and plays a crucial role in pain transmission.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos Opioides/farmacologia , Dor/metabolismo , Proteínas/metabolismo , Medula Espinal/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Analgésicos Opioides/efeitos adversos , Animais , Encéfalo/patologia , Células COS , Chlorocebus aethiops , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Peptídeos Opioides/efeitos adversos , Dor/genética , Proteínas/agonistas , Proteínas/genética , Medula Espinal/patologia , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Transmissão Sináptica/genética
5.
Front Genet ; 14: 1107787, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37007968

RESUMO

Tenascin-X (TNX) is an extracellular matrix glycoprotein for which a deficiency results in a recessive form of classical-like Ehlers-Danlos syndrome (clEDS), a heritable connective tissue disorder with hyperextensible skin without atrophic scarring, joint hypermobility, and easy bruising. Notably, patients with clEDS also suffer from not only chronic joint pain and chronic myalgia but also neurological abnormalities such as peripheral paresthesia and axonal polyneuropathy with high frequency. By using TNX-deficient (Tnxb -/-) mice, well-known as a model animal of clEDS, we recently showed that Tnxb -/- mice exhibit hypersensitivity to chemical stimuli and the development of mechanical allodynia due to the hypersensitization of myelinated A-fibers and activation of the spinal dorsal horn. Pain also occurs in other types of EDS. First, we review the underlying molecular mechanisms of pain in EDS, especially that in clEDS. In addition, the roles of TNX as a tumor suppressor protein in cancer progression have been reported. Recent in silico large-scale database analyses have shown that TNX is downregulated in various tumor tissues and that high expression of TNX in tumor cells has a good prognosis. We describe what is so far known about TNX as a tumor suppressor protein. Furthermore, some patients with clEDS show delayed wound healing. Tnxb -/- mice also exhibit impairment of epithelial wound healing in corneas. TNX is also involved in liver fibrosis. We address the molecular mechanism for the induction of COL1A1 by the expression of both a peptide derived from the fibrinogen-related domain of TNX and integrin α11.

6.
Sci Rep ; 13(1): 18490, 2023 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-37898719

RESUMO

Deficiency of an extracellular matrix glycoprotein tenascin-X (TNX) leads to a human heritable disorder Ehlers-Danlos syndrome, and TNX-deficient patients complain of chronic joint pain, myalgia, paresthesia, and axonal polyneuropathy. We previously reported that TNX-deficient (Tnxb-/-) mice exhibit mechanical allodynia and hypersensitivity to myelinated A-fibers. Here, we investigated the pain response of Tnxb-/- mice using pharmacological silencing of A-fibers with co-injection of N-(2,6-Dimethylphenylcarbamoylmethyl) triethylammonium bromide (QX-314), a membrane-impermeable lidocaine analog, plus flagellin, a toll-like receptor 5 (TLR5) ligand. Intraplantar co-injection of QX-314 and flagellin significantly increased the paw withdrawal threshold to transcutaneous sine wave stimuli at frequencies of 250 Hz (Aδ fiber responses) and 2000 Hz (Aß fiber responses), but not 5 Hz (C fiber responses) in wild-type mice. The QX-314 plus flagellin-induced silencing of Aδ- and Aß-fibers was also observed in Tnxb-/- mice. Co-injection of QX-314 and flagellin significantly inhibited the mechanical allodynia and neuronal activation of the spinal dorsal horn in Tnxb-/- mice. Interestingly, QX-314 alone inhibited the mechanical allodynia in Tnxb-/- mice, and it increased the paw withdrawal threshold to stimuli at frequencies of 250 Hz and 2000 Hz in Tnxb-/- mice, but not in wild-type mice. The inhibition of mechanical allodynia induced by QX-314 alone was blocked by intraplantar injection of a TLR5 antagonist TH1020 in Tnxb-/- mice. These results suggest that mechanical allodynia due to TNX deficiency is caused by the hypersensitivity of Aδ- and Aß-fibers, and it is induced by constitutive activation of TLR5.


Assuntos
Síndrome de Ehlers-Danlos , Hiperalgesia , Animais , Humanos , Camundongos , Síndrome de Ehlers-Danlos/complicações , Síndrome de Ehlers-Danlos/genética , Matriz Extracelular , Flagelina , Hiperalgesia/genética , Hiperalgesia/complicações , Fibras Nervosas Amielínicas , Tenascina/genética , Receptor 5 Toll-Like
7.
Glia ; 59(2): 208-18, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21125641

RESUMO

Neuropathic pain produced by damage to or dysfunction of the nervous system is a common and severely disabling state that affects millions of people worldwide. Recent evidence indicates that activated microglia are key cellular intermediaries in the pathogenesis of neuropathic pain and that ATP serves as the mediator. However, the in vivo mechanism underlying the retention of activated microglia in the injured region has not yet been completely elucidated. Prostaglandin E(2) (PGE(2)) is the principal proinflammatory prostanoid and plays versatile roles by acting via four PGE receptor subtypes, EP1-EP4. In the present study, we investigated the role of PGE(2) in spinal microglial activation in relation to neuropathic pain by using genetic and pharmacological methods. Mice deficient in microsomal prostaglandin E synthase-1 impaired the activation of microglia and the NMDA-nitric oxide (NO) cascade in spinal neurons in the dorsal horn and did not exhibit mechanical allodynia after peripheral nerve injury. The intrathecal injection of indomethacin, a nonsteroidal anti-inflammatory drug, ONO-8713, a selective EP1 antagonist, or 7-nitroindole, a neuronal NO synthase inhibitor, attenuated mechanical allodynia and the increase in activated microglia observed in the established neuropathic-pain state. We further demonstrated that ATP-induced microglial migration was blocked in vitro by PGE(2) via EP2 and by S-nitrosoglutathione, an NO donor. Taken together, the present study suggests that PGE(2) participated in the maintenance of neuropathic pain in vivo not only by activating spinal neurons, but also by retaining microglia in the central terminals of primary afferent fibers via EP2 subtype and via EP1-mediated NO production.


Assuntos
Movimento Celular/fisiologia , Dinoprostona/metabolismo , Microglia/fisiologia , Neuralgia/metabolismo , Neuralgia/patologia , Medula Espinal/patologia , Trifosfato de Adenosina/farmacologia , Animais , Movimento Celular/genética , Córtex Cerebral/citologia , Cinamatos/farmacologia , Cinamatos/uso terapêutico , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Indazóis/farmacologia , Indazóis/uso terapêutico , Oxirredutases Intramoleculares/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Microglia/efeitos dos fármacos , Neuralgia/complicações , Neuralgia/tratamento farmacológico , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Prostaglandina-E Sintases , S-Nitrosoglutationa/farmacologia , Medula Espinal/efeitos dos fármacos , Nervos Espinhais/lesões
8.
Biomed Rep ; 14(2): 21, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33335727

RESUMO

Nucleophosmin 1 (NPM1) primarily localizes to the nucleus and is passively released into the extracellular milieu by necrotic or damaged cells, or is secreted by monocytes and macrophages. Extracellular NPM1 acts as a potent inflammatory stimulator by promoting cytokine production [e.g., tumor necrosis factor-α (TNF-α)], which suggests that NPM1 acts as a damage-associated molecular pattern. However, the receptor of NPM1 is unknown. Evidence indicates that DAMPs, which include high mobility group box 1 and histones, may bind Toll-like receptors (TLRs). In the present study, it was shown that NPM1 signaling was mediated via the TLR4 pathway, which suggests that TLR4 is an NPM1 receptor. TLR4 binds myeloid differentiation protein-2 (MD-2), which is essential for intracellular signaling. Furthermore, the TLR4 antagonist, LPS-Rhodobacter sphaeroides (an MD-2 antagonist) and TAK-242 (a TLR4 signaling inhibitor) significantly inhibited NPM1-induced TNF-α production by differentiated THP-1 cells as well as reducing ERK1/2 activation. Far-western blot analysis revealed that NPM1 directly bound MD-2. Thus, the results of the present study provide compelling evidence that TLR4 binds NPM1, and it is hypothesized that inhibiting NPM1 activity may serve as a novel strategy for treating TLR4-related diseases.

9.
Eur J Neurosci ; 32(5): 798-810, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20722721

RESUMO

Ca(2+) /calmodulin-dependent protein kinase II (CaMKII) is a key mediator of long-term potentiation (LTP), which can be triggered by N-methyl-d-aspartate (NMDA) receptor-mediated Ca(2+) influx. We previously demonstrated that Fyn kinase-mediated phosphorylation of NR2B subunits of NMDA receptors at Tyr1472 in the dorsal horn was involved in a neuropathic pain state even 1 week after nerve injury. Here we show that Y1472F-KI mice with a knock-in mutation of the Tyr1472 site to phenylalanine did not exhibit neuropathic pain induced by L5 spinal nerve transection, whereas they did retain normal nociceptive responses and induction of inflammatory pain. Phosphorylation of NR2B at Tyr1472 was only impaired in the spinal cord of Y1472F-KI mice among the major phosphorylation sites. There was no difference in the Ca(2+) response to glutamate and sensitivity to NMDA receptor antagonists between naive wild-type and Y1472F-KI mice, and the Ca(2+) response to glutamate was attenuated in the Y1472F-KI mice after nerve injury. Autophosphorylation of CaMKII at Thr286 was markedly impaired in Y1472F-KI mice after nerve injury, but there was no difference in phosphorylation of CaMKII at Thr305 or protein kinase Cγ at Thr674, and activation of neuronal nitric oxide synthase and microglia in the superficial layer of spinal cord between wild-type and Y1472F-KI mice after the operation. These results demonstrate that the attenuation of neuropathic pain is caused by the impaired NMDA receptor-mediated CaMKII signaling in Y1472F-KI mice, and suggest that autophosphorylation of CaMKII at Thr286 plays a central part not only in LTP, but also in persistent neuropathic pain.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Neuralgia/fisiopatologia , Dor/fisiopatologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Ácido Glutâmico/farmacologia , Inflamação/genética , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Neuralgia/genética , Óxido Nítrico Sintase Tipo I/biossíntese , Fosforilação/genética , Proteína Quinase C/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Nervos Espinhais/lesões , Nervos Espinhais/fisiopatologia
10.
Sci Rep ; 10(1): 6569, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32300146

RESUMO

Tenascin-X (TNX) is a member of the extracellular matrix glycoprotein tenascin family, and TNX deficiency leads to Ehlers-Danlos syndrome, a heritable human disorder characterized mostly by skin hyperextensibility, joint hypermobility, and easy bruising. TNX-deficient patients complain of chronic joint pain, myalgia, paresthesia, and axonal polyneuropathy. However, the molecular mechanisms by which TNX deficiency complicates pain are unknown. Here, we examined the nociceptive behavioral responses of TNX-deficient mice. Compared with wild-type mice, TNX-deficient mice exhibited mechanical allodynia but not thermal hyperalgesia. TNX deficiency also increased pain sensitivity to chemical stimuli and aggravated early inflammatory pain elicited by formalin. TNX-deficient mice were significantly hypersensitive to transcutaneous sine wave stimuli at frequencies of 250 Hz (Aδ fiber responses) and 2000 Hz (Aß fiber responses), but not to stimuli at frequency of 5 Hz (C fiber responses). In addition, the phosphorylation levels of extracellular signal-related kinase, an active neuronal marker, and the activity of NADPH-diaphorase, a neuronal nitric oxide activation marker, were enhanced in the spinal dorsal horns of TNX-deficient mice. These results suggest that TNX deficiency contributes to the development of mechanical allodynia and hypersensitivity to chemical stimuli, and it induces hypersensitization of myelinated A fibers and activation of the spinal dorsal horn.


Assuntos
Síndrome de Ehlers-Danlos/complicações , Hiperalgesia/complicações , Tenascina/deficiência , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Formaldeído , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Dor/complicações , Dor/patologia , Dor/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Corno Dorsal da Medula Espinal/efeitos dos fármacos , Corno Dorsal da Medula Espinal/patologia , Corno Dorsal da Medula Espinal/fisiopatologia , Tenascina/genética , Tenascina/metabolismo
11.
Mol Pain ; 5: 58, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19785772

RESUMO

BACKGROUND: The role of the diffusible messenger nitric oxide (NO) in the regulation of pain transmission is still a debate of matter, pro-nociceptive and/or anti-nociceptive. S-Nitrosylation, the reversible post-translational modification of selective cysteine residues in proteins, has emerged as an important mechanism by which NO acts as a signaling molecule. The occurrence of S-nitrosylation in the spinal cord and its targets that may modulate pain transmission remain unclarified. The "biotin-switch" method and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry were employed for identifying S-nitrosylated proteins. RESULTS: Here we show that actin was a major protein S-nitrosylated in the spinal cord by the NO donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP). Interestingly, actin was S-nitrosylated, more in the S2 fraction than in the P2 fraction of the spinal homogenate. Treatment of PC12 cells with SNAP caused rapid S-nitrosylation of actin and inhibited dopamine release from the cells. Just like cytochalasin B, which depolymerizes actin, SNAP decreased the amount of filamentous actin cytoskeleton just beneath the membrane. The inhibition of dopamine release was not attenuated by inhibitors of soluble guanylyl cyclase and cGMP-dependent protein kinase. CONCLUSION: The present study demonstrates that actin is a major S-nitrosylated protein in the spinal cord and suggests that NO directly regulates neurotransmitter release by S-nitrosylation in addition to the well-known phosphorylation by cGMP-dependent protein kinase.


Assuntos
Actinas/metabolismo , Neurotransmissores/metabolismo , Óxido Nítrico/farmacologia , S-Nitroso-N-Acetilpenicilamina/metabolismo , Sequência de Aminoácidos , Animais , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Dopamina/metabolismo , Camundongos , Dados de Sequência Molecular , Células PC12 , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Proteínas/química , Ratos , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Medula Espinal/efeitos dos fármacos , Medula Espinal/enzimologia
12.
J Neurochem ; 105(6): 2271-85, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18331476

RESUMO

In the central nervous system, the activation of neuronal nitric oxide synthase (nNOS) is closely associated with activation of NMDA receptor, and trafficking of nNOS may be a prerequisite for efficient NO production at synapses. We recently demonstrated that pituitary adenylate cyclase activating polypeptide (PACAP) and NMDA synergistically caused the translocation of nNOS to the membrane and stimulated NO production in PC12 (pheochromocytoma) cells. However, the mechanisms responsible for trafficking and activation of nNOS are largely unknown. To address these issues, here we constructed a yellow fluorescent protein (YFP)-tagged nNOS N-terminal (1-299 a.a.) mutant, nNOSNT-YFP, and visualized its translocation in PC12 cells stably expressing it. PACAP enhanced the translocation synergistically with NMDA in a time- and concentration-dependent manner. The translocation was blocked by inhibitors of protein kinase A (PKA), protein kinase C (PKC), and Src kinase; and the effect of PACAP could be replaced with PKA and PKC activators. The beta-finger region in the PSD-95/disc large/zonula occludens-1 domain of nNOS was required for the translocation of nNOS and its interaction with post-synaptic density-95 (PSD-95), and NO formation was attenuated by dominant negative nNOSNT-YFP. These results demonstrate that PACAP stimulated nNOS translocation mediated by PKA and PKC via PAC(1)-receptor (a PACAP receptor) and suggest cross-talk between PACAP and NMDA for nNOS activation by Src-dependent phosphorylation of NMDA receptors.


Assuntos
Membrana Celular/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Transdução de Sinais/fisiologia , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , N-Metilaspartato/química , N-Metilaspartato/fisiologia , Óxido Nítrico Sintase Tipo I/química , Células PC12 , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/química , Proteína Quinase C/química , Proteína Quinase C/fisiologia , Transporte Proteico/fisiologia , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/química , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia
13.
Eur J Neurosci ; 27(12): 3161-70, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18598260

RESUMO

Central sensitization, similar to long-term potentiation in the hippocampus, refers to the increased synaptic efficacy established in somatosensory neurons in the dorsal horn of the spinal cord following tissue injury or nerve damage. In the course of inflammation, many proteins including glutamate receptors are assumed to be dynamically reorganized in the postsynaptic density (PSD) and involved in persistent pain. Mechanical hyperalgesia induced by intraplantar injection of complete Freund's adjuvant (CFA) was inhibited at 4 h, but not at 24 h, by indomethacin, an inhibitor of prostanoid synthesis. To elucidate the nature of the molecule(s) involved in the late phase of inflammatory pain, we analysed the PSD fraction prepared from the lumbar spinal cord of rats before and 24 h after CFA injection by conducting two-dimensional differential gel electrophoresis. N-ethylmaleimide-sensitive fusion protein (NSF) was identified as a downregulated protein in the PSD by MALDI-TOF MS and immunoblotting. Concomitant with the decrease in NSF, GluR2 and GluR3 were decreased and GluR1 was conversely increased in the PSD fraction 24 h after CFA injection. In vivo patch-clamp recordings of rats 24 h after CFA injection showed that excitatory postsynaptic currents of dorsal horn neurons evoked by pinch stimuli to inflamed skin were inwardly rectified and inhibited by 60% by philanthotoxin-433, a selective inhibitor of the Ca2+-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor. These results suggest that peripheral inflammation gives rise to central sensitization in the spinal cord through subunit composition switch of AMPA receptors in the late phase.


Assuntos
Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Neuralgia/metabolismo , Neurite (Inflamação)/metabolismo , Receptores de AMPA/metabolismo , Medula Espinal/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anticorpos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Adjuvante de Freund , Indometacina/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Proteínas Sensíveis a N-Etilmaleimida/imunologia , Neuralgia/tratamento farmacológico , Neuralgia/imunologia , Neurite (Inflamação)/tratamento farmacológico , Neurite (Inflamação)/imunologia , Neurônios Aferentes/metabolismo , Técnicas de Cultura de Órgãos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Técnicas de Patch-Clamp , Coelhos , Ratos , Ratos Wistar , Receptores de AMPA/imunologia , Medula Espinal/imunologia , Sinapses/metabolismo , Fatores de Tempo
14.
Neuropharmacology ; 52(5): 1318-25, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17350656

RESUMO

Nociceptin/orphanin FQ (N/OFQ) was earlier shown to be involved in the maintenance of neuropathic pain by activating neuronal nitric oxide synthase (nNOS). We recently established an ex vivo system to elucidate biochemical and molecular mechanisms for nNOS activation by the use of a combination of isolated intact spinal cord preparations and NADPH-diaphorase histochemistry. Here we examined the N/OFQ signal pathways coupled to nNOS activation in the spinal cord by using this ex vivo system. N/OFQ enhanced nNOS activity in the superficial layer of the spinal cord, as assessed by NADPH-diaphorase histochemistry, in a time- and dose-dependent manner. The maximum effect was observed at 3-10 nM. The N/OFQ-stimulated nNOS activity was inhibited by NMDA receptor antagonists MK-801 and D-AP5, but not by the NR2B-selective antagonist CP-101,606; and the stimulated activity was observed in NR2D(-/-) mice, but not in NR2A(-/-) or NR2A(-/-)/NR2D(-/-) mice. N/OFQ receptor antagonists attenuated the nNOS activity stimulated by N/OFQ, but not that by NMDA. Furthermore, the potentiation of nNOS by N/OFQ was inhibited by calphostin C and Ro 31-8220, PP2, and KN-62, but not by H-89. These results suggest that N/OFQ stimulated nNOS activity by a biochemical cascade initiated by activation of NMDA receptors containing NR2A.


Assuntos
Óxido Nítrico Sintase Tipo I/metabolismo , Peptídeos Opioides/farmacologia , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/enzimologia , Animais , AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Desidrogenase/metabolismo , Antagonistas de Entorpecentes , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores Opioides , Receptor de Nociceptina , Nociceptina
15.
Invest Ophthalmol Vis Sci ; 48(1): 455-63, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17197567

RESUMO

PURPOSE: A prior study showed inactivation of ornithine-delta-aminotransferase (OAT)-deficient human retinal pigment epithelial (RPE) cells by a specific irreversible inhibitor (5-fluoromethylornithine; 5-FMO) leading to cell death, in an in vitro model of gyrate atrophy (GA) of the choroid and retina. In the present study, the cytotoxicity of metabolites of ornithine, especially spermine, in RPE cells was investigated, to clarify the mechanism of ornithine cytotoxicity in RPE cells. METHODS: RPE cells were incubated with ornithine or compounds involved in ornithine metabolic pathways. The effects on RPE cell viability and proliferative activity were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric and [(3)H]thymidine incorporation assays. Incorporation of spermine into RPE cells was examined by using [(14)C]spermine and dansyl-spermine. To assess spermine-induced RPE cell death, cells were double stained with annexin V and propidium iodide and subjected to flow cytometry. RESULTS: Ornithine, arginine, glutamate, proline, creatine, glycine, and putrescine exhibited no effects on the viability and proliferative activities of RPE cells, whereas spermidine and spermine (10 mM) inhibited [(3)H]thymidine incorporation by 13% and 89%, respectively. The inhibition of [(3)H]thymidine incorporation by spermine was dose dependent and was observed as early as 4 hours after addition. Further, spermine was incorporated and accumulated in the perinuclear region of RPE cells. Apoptotic RPE cell death was induced by spermine in a dose-dependent manner. CONCLUSIONS: The present results demonstrated that excessive spermine is cytotoxic to RPE cells and suggest that metabolites of ornithine, especially spermine, may be involved in the mechanism of RPE degeneration in GA.


Assuntos
Apoptose/efeitos dos fármacos , Epitélio Pigmentado Ocular/efeitos dos fármacos , Espermina/toxicidade , Animais , Anexina A5/metabolismo , Bovinos , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Colorimetria , DNA/biossíntese , Relação Dose-Resposta a Droga , Citometria de Fluxo , Microscopia Confocal , Ornitina/toxicidade , Epitélio Pigmentado Ocular/patologia , Propídio/metabolismo , Sais de Tetrazólio , Tiazóis , Timidina/metabolismo
16.
Invest Ophthalmol Vis Sci ; 48(1): 464-71, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17197568

RESUMO

PURPOSE: A prior report showed ornithine cytotoxicity in ornithine-delta-aminotransferase (OAT)-deficient human retinal pigment epithelial (RPE) cells in an in vitro model of gyrate atrophy of the choroid and retina. This study was intended to clarify the mechanism of ornithine cytotoxicity and to determine the responsible amino acid transporters. METHODS: The mRNA expression of amino acid transporters in human telomerase reverse transcriptase (hTERT)-RPE cells was examined by reverse transcription polymerase chain reaction (RT-PCR) and Northern blot analysis. Carrier-mediated ornithine transport via the L-type amino acid transporter (LAT)1, LAT2, cationic amino acid transporter (CAT)-1, and y(+)LAT2 systems was evaluated by short interfering (si)RNA-mediated gene silencing. The cytoprotective effect of CAT-1-specific siRNA on ornithine cytotoxicity was measured using quantitative analysis of cellular adenosine triphosphate (ATP) at 24 hours after treatment with ornithine in OAT-deficient RPE cells. RESULTS: LAT1, LAT2, CAT-1, and y(+)LAT2 mRNA expression was detected by Northern blot analysis, whereas RT-PCR revealed that LAT1, LAT2, y(+)LAT1, y(+)LAT2, CAT-1, and b(0,+)AT mRNAs were expressed together with the heterodimeric glycoproteins 4F2hc and rBAT in hTERT-RPE cells. l-[(14)C]ornithine uptake in hTERT-RPE cells was decreased by 46.6% and 22.0% by CAT-1 and y(+)LAT2 siRNA, respectively, whereas LAT1 and LAT2 siRNA had no significant effect. Further, CAT-1 silencing by siRNA reduced ornithine cytotoxicity in OAT-deficient RPE cells. CONCLUSIONS: The results suggest that ornithine transport via CAT-1 may play a crucial role in ornithine cytotoxicity in hTERT-RPE cells. Reduction of the ornithine transport via CAT-1 may be a new target for treatment of gyrate atrophy.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/metabolismo , Ornitina/metabolismo , Ornitina/toxicidade , Epitélio Pigmentado Ocular/metabolismo , Trifosfato de Adenosina/metabolismo , Transporte Biológico , Northern Blotting , Transportador 1 de Aminoácidos Catiônicos/genética , Células Cultivadas , Inativação Gênica , Humanos , Leucina/metabolismo , Ornitina-Oxo-Ácido Transaminase/metabolismo , Epitélio Pigmentado Ocular/efeitos dos fármacos , Plasmídeos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Neurosci ; 24(33): 7283-91, 2004 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-15317855

RESUMO

The prolonged sensitization of pain transmission after nerve injury by increasing excitability of spinal neurons and thereby promoting repair is an adaptive response of the body. The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is widely distributed in the nervous system and implicated in neurotransmission, neural plasticity, and neurotrophic actions. Although PACAP is distributed in the spinal cord and dorsal root ganglia, a role of PACAP in pain responses remains essentially unknown. Here we show that mice lacking the PACAP gene (PACAP-/-) did not exhibit inflammatory pain induced by intraplantar injection of carrageenan or neuropathic pain induced by L5 spinal nerve transection, whereas they did retain normal nociceptive responses. Intrathecal administration of NMDA induced mechanical allodynia in wild-type mice, but not in PACAP-/- mice. The NMDA-induced allodynia in PACAP-/- mice was reproduced by simultaneous intrathecal injection of PACAP with NMDA. Concomitant with the increase in PACAP immunoreactivity after nerve injury, NADPH-dependent nitric oxide synthase (NOS) activity visualized by NADPH diaphorase histochemistry markedly increased in the superficial layer of the spinal cord of wild-type mice, which was not observed in PACAP-/- mice. Simultaneous addition of PACAP and NMDA caused translocation of neuronal NOS from the cytosol to the membrane and stimulated NO production in vitro. These results demonstrate that PACAP might promote the functional coupling of neuronal NOS to NMDA receptors for both inflammatory and neuropathic pain to occur.


Assuntos
Neuropeptídeos/fisiologia , Dor/etiologia , Medula Espinal/metabolismo , Animais , Membrana Celular/enzimologia , Inflamação/etiologia , Camundongos , Camundongos Knockout , N-Metilaspartato/toxicidade , Neuropeptídeos/genética , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Dor/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Transporte Proteico
19.
Curr Pharm Des ; 21(7): 868-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25345611

RESUMO

A neuropeptide nociceptin or orphanin FQ (N/OFQ) is an endogenous ligand for the orphan opioid receptor-like receptor. During studies on the analysis of the precursor of N/OFQ, we identified a novel neuropeptide produced from the same precursor and named it "nocistatin (NST)". Intrathecal (i.t.) administration of N/OFQ induces pain responses including touch-evoked allodynia and thermal hyperalgesia, and simultaneous administration of NST blocks the allodynia and hyperalgesia induced by N/OFQ. In the years since these discoveries, N/OFQ has been shown to be involved in a wide range of pharmacological activities, such as relaying pain perception in peripheral tissues, to the central nervous system, and NST was shown to have opposite effects on various central functions evoked by N/OFQ. Pharmacological characterization using various neurotransmitter agents, agonists, antagonists and knockout mice in vivo; electrophysiological and immunohistological analysis ex vivo; and molecular cloning using affinity chromatography of high-performance affinity nanobeads; and protein processing measurement using bioluminescence resonance energy transfer (BRET) in vitro have generated new insights into pain transmission regulated by NST and N/OFQ. This review focuses on the molecular and cellular mechanisms of pain transmission regulated by NST.


Assuntos
Peptídeos Opioides/genética , Peptídeos Opioides/metabolismo , Dor/genética , Dor/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Nociceptividade/fisiologia , Medição da Dor/métodos , Sinapses/metabolismo , Fatores de Tempo
20.
Vitam Horm ; 97: 147-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25677771

RESUMO

Nociceptin/orphanin FQ (N/OFQ) and nocistatin (NST) are neuropeptides produced from the same precursor protein. N/OFQ is involved in a broad range of central functions including pain, learning, memory, anxiety, and feeding. However, NST has opposite effects on various central functions evoked by N/OFQ. The regulation of their receptors may be important for these opposite functions of NST and N/OFQ. Although N/OFQ binds to a specific N/OFQ receptor, the target molecule of NST remains unclear. Some biological effects of NST are mediated by a G protein-coupled receptor. Furthermore, using high-performance affinity nanobeads, we recently identified a 4-nitrophenylphosphatase domain and nonneuronal SNAP25-like protein homolog 1 (NIPSNAP1) as a protein that interacts with NST in the mouse spinal cord. The inhibition of N/OFQ-evoked tactile pain allodynia by NST is mediated by NIPSNAP1. This review focuses on the molecular mechanisms of pain regulation by the target molecules of NST including a G protein-coupled receptor and NIPSNAP1.


Assuntos
Drogas em Investigação/farmacologia , Antagonistas de Entorpecentes/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Peptídeos Opioides/antagonistas & inibidores , Proteínas/metabolismo , Receptores Opioides/metabolismo , Animais , Drogas em Investigação/química , Drogas em Investigação/metabolismo , Drogas em Investigação/uso terapêutico , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/uso terapêutico , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/metabolismo , Peptídeos Opioides/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Proteínas/agonistas , Proteínas/antagonistas & inibidores , Receptores Opioides/agonistas , Receptores Opioides/química , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA