Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Physiol ; 595(11): 3497-3514, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28402052

RESUMO

KEY POINTS: A subpopulation of retinal ganglion cells expresses the neuropeptide vasopressin. These retinal ganglion cells project predominately to our biological clock, the suprachiasmatic nucleus (SCN). Light-induced vasopressin release enhances the responses of SCN neurons to light. It also enhances expression of genes involved in photo-entrainment of biological rhythms. ABSTRACT: In all animals, the transition between night and day engages a host of physiological and behavioural rhythms. These rhythms depend not on the rods and cones of the retina, but on retinal ganglion cells (RGCs) that detect the ambient light level in the environment. These project to the suprachiasmatic nucleus (SCN) of the hypothalamus to entrain circadian rhythms that are generated within the SCN. The neuropeptide vasopressin has an important role in this entrainment. Many SCN neurons express vasopressin, and it has been assumed that the role of vasopressin in the SCN reflects the activity of these cells. Here we show that vasopressin is also expressed in many retinal cells that project to the SCN. Light-evoked vasopressin release contributes to the responses of SCN neurons to light, and enhances expression of the immediate early gene c-fos in the SCN, which is involved in photic entrainment of circadian rhythms.


Assuntos
Luz , Células Ganglionares da Retina/metabolismo , Núcleo Supraquiasmático/metabolismo , Vasopressinas/metabolismo , Animais , Ritmo Circadiano , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Núcleo Supraquiasmático/fisiologia
2.
Nat Med ; 12(11): 1269-77, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17057711

RESUMO

We report that GTP cyclohydrolase (GCH1), the rate-limiting enzyme for tetrahydrobiopterin (BH4) synthesis, is a key modulator of peripheral neuropathic and inflammatory pain. BH4 is an essential cofactor for catecholamine, serotonin and nitric oxide production. After axonal injury, concentrations of BH4 rose in primary sensory neurons, owing to upregulation of GCH1. After peripheral inflammation, BH4 also increased in dorsal root ganglia (DRGs), owing to enhanced GCH1 enzyme activity. Inhibiting this de novo BH4 synthesis in rats attenuated neuropathic and inflammatory pain and prevented nerve injury-evoked excess nitric oxide production in the DRG, whereas administering BH4 intrathecally exacerbated pain. In humans, a haplotype of the GCH1 gene (population frequency 15.4%) was significantly associated with less pain following diskectomy for persistent radicular low back pain. Healthy individuals homozygous for this haplotype exhibited reduced experimental pain sensitivity, and forskolin-stimulated immortalized leukocytes from haplotype carriers upregulated GCH1 less than did controls. BH4 is therefore an intrinsic regulator of pain sensitivity and chronicity, and the GTP cyclohydrolase haplotype is a marker for these traits.


Assuntos
Biopterinas/análogos & derivados , GTP Cicloidrolase/fisiologia , Limiar da Dor/fisiologia , Dor/fisiopatologia , Adulto , Oxirredutases do Álcool/antagonistas & inibidores , Animais , Biopterinas/fisiologia , Inibidores Enzimáticos/farmacologia , GTP Cicloidrolase/antagonistas & inibidores , Haplótipos , Humanos , Manejo da Dor , Estudos Prospectivos , Ratos
3.
EMBO Rep ; 11(6): 473-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20467438

RESUMO

Sensitization to inflammatory pain is a pathological form of neuronal plasticity that is poorly understood and treated. Here we examine the role of the SH3 domain of postsynaptic density 95 (PSD95) by using mice that carry a single amino-acid substitution in the polyproline-binding site. Testing multiple forms of plasticity we found sensitization to inflammation was specifically attenuated. The inflammatory response required recruitment of phosphatidylinositol-3-kinase-C2alpha to the SH3-binding site of PSD95. In wild-type mice, wortmannin or peptide competition attenuated the sensitization. These results show that different types of behavioural plasticity are mediated by specific domains of PSD95 and suggest novel therapeutic avenues for reducing inflammatory pain.


Assuntos
Inflamação/complicações , Inflamação/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Dor/complicações , Dor/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Domínios de Homologia de src , Animais , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases , Hipocampo/enzimologia , Hipocampo/patologia , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Plasticidade Neuronal , Mutação Puntual/genética , Ligação Proteica , Relação Estrutura-Atividade , Sinapses/enzimologia
4.
Brain ; 133(9): 2519-27, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20724292

RESUMO

Not all patients with nerve injury develop neuropathic pain. The extent of nerve damage and age at the time of injury are two of the few risk factors identified to date. In addition, preclinical studies show that neuropathic pain variance is heritable. To define such factors further, we performed a large-scale gene profiling experiment which plotted global expression changes in the rat dorsal root ganglion in three peripheral neuropathic pain models. This resulted in the discovery that the potassium channel alpha subunit KCNS1, involved in neuronal excitability, is constitutively expressed in sensory neurons and markedly downregulated following nerve injury. KCNS1 was then characterized by an unbiased network analysis as a putative pain gene, a result confirmed by single nucleotide polymorphism association studies in humans. A common amino acid changing allele, the 'valine risk allele', was significantly associated with higher pain scores in five of six independent patient cohorts assayed (total of 1359 subjects). Risk allele prevalence is high, with 18-22% of the population homozygous, and an additional 50% heterozygous. At lower levels of nerve damage (lumbar back pain with disc herniation) association with greater pain outcome in homozygote patients is P = 0.003, increasing to P = 0.0001 for higher levels of nerve injury (limb amputation). The combined P-value for pain association in all six cohorts tested is 1.14 E-08. The risk profile of this marker is additive: two copies confer the most, one intermediate and none the least risk. Relative degrees of enhanced risk vary between cohorts, but for patients with lumbar back pain, they range between 2- and 3-fold. Although work still remains to define the potential role of this protein in the pathogenic process, here we present the KCNS1 allele rs734784 as one of the first prognostic indicators of chronic pain risk. Screening for this allele could help define those individuals prone to a transition to persistent pain, and thus requiring therapeutic strategies or lifestyle changes that minimize nerve injury.


Assuntos
Canal de Potássio Kv1.1/genética , Dor/genética , Polimorfismo Genético/genética , Valina/genética , Animais , Doença Crônica , Estudos de Coortes , Compreensão , Biologia Computacional/métodos , Comparação Transcultural , Modelos Animais de Doenças , Feminino , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Desequilíbrio de Ligação , Masculino , Proteínas de Neurofilamentos , Neuropeptídeos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Dor/etiologia , Ratos
5.
J Neuroendocrinol ; 33(5): e12970, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33851744

RESUMO

Oxytocin is primarily synthesised in the brain and is widely known for its role in lactation and parturition after being released into the blood from the posterior pituitary gland. Nevertheless, peripheral tissues have also been reported to express oxytocin. Using systemic injection of a recombinant adeno-associated virus vector, we investigated the expression of the green fluorescent protein Venus under the control of the oxytocin promoter in the gastrointestinal tract, pancreas and testes of adult rats. Here, we confirm that the vector infects oxytocin neurones of the enteric nervous system in ganglia of the myenteric and submucosal plexuses. Venus was detected in 25%-60% of the ganglia in the myenteric and submucosal plexuses identified by co-staining with the neuronal marker PGP9.5. Oxytocin expression was also detected in the islets of Langerhans in the pancreas and the Leydig cells of the testes. Our data illustrate that peripheral administration of the viral vector represents a powerful method for selectively labelling oxytocin-producing cells outside the brain.


Assuntos
Sistema Nervoso Entérico/metabolismo , Neurônios/metabolismo , Ocitocina/metabolismo , Animais , Trato Gastrointestinal/metabolismo , Masculino , Pâncreas/metabolismo , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Testículo/metabolismo
6.
Neuron ; 50(2): 277-89, 2006 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-16630838

RESUMO

TRPA1, a member of the transient receptor potential (TRP) family of ion channels, is expressed by dorsal root ganglion neurons and by cells of the inner ear, where it has proposed roles in sensing sound, painful cold, and irritating chemicals. To test the in vivo roles of TRPA1, we generated a mouse in which the essential exons required for proper function of the Trpa1 gene were deleted. Knockout mice display behavioral deficits in response to mustard oil, to cold ( approximately 0 degrees C), and to punctate mechanical stimuli. These mice have a normal startle reflex to loud noise, a normal sense of balance, a normal auditory brainstem response, and normal transduction currents in vestibular hair cells. TRPA1 is apparently not essential for hair-cell transduction but contributes to the transduction of mechanical, cold, and chemical stimuli in nociceptor sensory neurons.


Assuntos
Mapeamento Encefálico , Células Ciliadas Auditivas/fisiologia , Mecanotransdução Celular/fisiologia , Dor/fisiopatologia , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Percepção Auditiva/fisiologia , Temperatura Baixa , Camundongos , Camundongos Knockout , Nociceptores/metabolismo , Estimulação Física , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética
7.
J Neuroendocrinol ; 31(12): e12806, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31677199

RESUMO

Physiological circadian rhythms are orchestrated by the hypothalamic suprachiasmatic nucleus (SCN). The activity of SCN cells is synchronised by environmental signals, including light information from retinal ganglion cells (RGCs). We recently described a population of vasopressin-expressing RGCs (VP-RGC) that send axonal projections to the SCN. To determine how these VP-RGCs influence the activity of cells in the SCN, we used optogenetic tools to specifically activate their axon terminals within the SCN. Rats were intravitreally injected with a recombinant adeno-associated virus to express the channelrhodopsin-2 and the red fluorescent protein mCherry under the vasopressin promoter (VP-ChR2mCherry). In vitro recordings in acute brain slices showed that approximately 30% of ventrolateral SCN cells responded to optogenetic stimulation with an increase in firing rate that progressively increased during the first 200 seconds of stimulation and which persisted after the end of stimulation. Finally, application of a vasopressin V1A receptor antagonist dampened the response to optogenetic stimulation. Our data suggest that optogenetic stimulation of VP-RGC axons within the SCN influences the activity of SCN cells in a vasopressin-dependent manner.


Assuntos
Neurônios/fisiologia , Optogenética , Células Ganglionares da Retina/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia , Vasopressinas/fisiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Channelrhodopsins/genética , Feminino , Masculino , Ratos , Ratos Transgênicos , Receptores de Vasopressinas/efeitos dos fármacos
8.
J Neurosci ; 27(32): 8699-708, 2007 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-17687047

RESUMO

Microarray expression profiles reveal substantial changes in gene expression in the ipsilateral dorsal horn of the spinal cord in response to three peripheral nerve injury models of neuropathic pain. However, only 54 of the 612 regulated genes are commonly expressed across all the neuropathic pain models. Many of the commonly regulated transcripts are immune related and include the complement components C1q, C3, and C4, which we find are expressed only by microglia. C1q and C4 are, moreover, the most strongly regulated of all 612 regulated genes. In addition, we find that the terminal complement component C5 and the C5a receptor (C5aR) are upregulated in spinal microglia after peripheral nerve injury. Mice null for C5 had reduced neuropathic pain sensitivity, excluding C3a as a pain effector. C6-deficient rats, which cannot form the membrane attack complex, have a normal neuropathic pain phenotype. However, C5a applied intrathecally produces a dose-dependent, slow-onset cold pain in naive animals. Furthermore, a C5aR peptide antagonist reduces cold allodynia in neuropathic pain models. We conclude that induction of the complement cascade in spinal cord microglia after peripheral nerve injury contributes to neuropathic pain through the release and action of the C5a anaphylatoxin peptide.


Assuntos
Anafilatoxinas/biossíntese , Complemento C5a/biossíntese , Microglia/metabolismo , Dor/metabolismo , Medula Espinal/metabolismo , Anafilatoxinas/genética , Anafilatoxinas/fisiologia , Animais , Células Cultivadas , Complemento C5a/genética , Complemento C5a/fisiologia , Regulação da Expressão Gênica/fisiologia , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Dor/genética , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptor da Anafilatoxina C5a , Receptores de Complemento/biossíntese , Receptores de Complemento/genética
9.
J Neurosci ; 26(50): 12852-60, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17167076

RESUMO

We used a mouse with deletion of exons 4, 5, and 6 of the SCN11A (sodium channel, voltage-gated, type XI, alpha) gene that encodes the voltage-gated sodium channel Na(v)1.9 to assess its contribution to pain. Na(v)1.9 is present in nociceptor sensory neurons that express TRPV1, bradykinin B2, and purinergic P2X3 receptors. In Na(v)1.9-/- mice, the non-inactivating persistent tetrodotoxin-resistant sodium TTXr-Per current is absent, whereas TTXr-Slow is unchanged. TTXs currents are unaffected by the mutation of Na(v)1.9. Pain hypersensitivity elicited by intraplantar administration of prostaglandin E2, bradykinin, interleukin-1beta, capsaicin, and P2X3 and P2Y receptor agonists, but not NGF, is either reduced or absent in Na(v)1.9-/- mice, whereas basal thermal and mechanical pain sensitivity is unchanged. Thermal, but not mechanical, hypersensitivity produced by peripheral inflammation (intraplanatar complete Freund's adjuvant) is substantially diminished in the null allele mutant mice, whereas hypersensitivity in two neuropathic pain models is unchanged in the Na(v)1.9-/- mice. Na(v)1.9 is, we conclude, an effector of the hypersensitivity produced by multiple inflammatory mediators on nociceptor peripheral terminals and therefore plays a key role in mediating peripheral sensitization.


Assuntos
Hiperalgesia/metabolismo , Neuropeptídeos/biossíntese , Nervos Periféricos/metabolismo , Canais de Sódio/biossíntese , Animais , Feminino , Hiperalgesia/genética , Inflamação/genética , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.9 , Neurônios Aferentes/metabolismo , Neuropeptídeos/genética , Dor/genética , Dor/metabolismo , Medição da Dor/métodos , Canais de Sódio/genética
10.
Mol Pain ; 3: 21, 2007 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-17672895

RESUMO

BACKGROUND: Clinical and experimental studies of neuropathic pain support the hypothesis that a functional coupling between postganglionic sympathetic efferent and sensory afferent fibers contributes to the pain. We investigated whether neuropathic pain-related behavior in the spared nerve injury (SNI) rat model is dependent on the sympathetic nervous system. RESULTS: Permanent chemical sympathectomy was achieved by daily injection of guanethidine (50 mg/kg s.c.) from age P8 to P21. SNI was performed at adulthood followed by 11 weeks of mechanical and thermal hypersensitivity testing. A significant but limited effect of the sympathectomy on SNI-induced pain sensitivity was observed. The effect was delayed and restricted to cold allodynia-like behavior: SNI-related cold scores were lower in the sympathectomized group compared to the control group at 8 and 11 weeks after the nerve injury but not before. Mechanical hypersensitivity tests (pinprick and von Frey hair threshold tests) showed no difference between groups during the study period. Concomitantly, pericellular tyrosine-hydroxylase immunoreactive basket structures were observed around dorsal root ganglia (DRG) neurons 8 weeks after SNI, but were absent at earlier time points after SNI and in sham operated controls. CONCLUSION: These results suggest that the early establishment of neuropathic pain-related behavior after distal nerve injury such as in the SNI model is mechanistically independent of the sympathetic system, whereas the system contributes to the maintenance, albeit after a delay of many weeks, of response to cold-related stimuli.


Assuntos
Neuralgia/fisiopatologia , Nervo Isquiático/fisiopatologia , Nervos Espinhais/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/enzimologia , Gânglios Espinais/patologia , Guanetidina/administração & dosagem , Guanetidina/farmacologia , Imuno-Histoquímica , Masculino , Neuralgia/metabolismo , Neuralgia/patologia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/lesões , Nervo Isquiático/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Nervos Espinhais/lesões , Nervos Espinhais/metabolismo , Simpatectomia/métodos , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
11.
J Neurosci ; 25(35): 7986-92, 2005 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-16135755

RESUMO

Bradykinin, an inflammatory mediator, sensitizes nociceptor peripheral terminals reducing pain threshold. We now show that the B2 kinin receptor is expressed in rat dorsal horn neurons and that bradykinin, a B2-specific agonist, augments AMPA- and NMDA-induced, and primary afferent-evoked EPSCs, and increases the frequency and amplitude of miniature EPSCs in superficial dorsal horn neurons in vitro. Administration of bradykinin to the spinal cord in vivo produces, moreover, an NMDA-dependent hyperalgesia. We also demonstrate that nociceptive inputs result in the production of bradykinin in the spinal cord and that an intrathecal B2-selective antagonist suppresses behavioral manifestations of central sensitization, an activity-dependent increase in glutamatergic synaptic efficacy. Primary afferent-evoked central sensitization is, in addition, reduced in B2 receptor knock-out mice. We conclude that bradykinin is released in the spinal cord in response to nociceptor inputs and acts as a synaptic neuromodulator, potentiating glutamatergic synaptic transmission to produce pain hypersensitivity.


Assuntos
Bradicinina/farmacologia , Hiperalgesia/induzido quimicamente , N-Metilaspartato/farmacologia , Medula Espinal/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Animais , Bradicinina/fisiologia , Sinergismo Farmacológico , Ácido Glutâmico/fisiologia , Hiperalgesia/fisiopatologia , Técnicas In Vitro , Camundongos , Camundongos Knockout , N-Metilaspartato/fisiologia , Dor/induzido quimicamente , Dor/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptor B2 da Bradicinina/agonistas , Receptor B2 da Bradicinina/fisiologia , Medula Espinal/fisiologia , Transmissão Sináptica/efeitos dos fármacos
12.
Neurosci Lett ; 395(3): 201-5, 2006 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-16332410

RESUMO

Early onset generalized dystonia is a dominantly inherited movement disorder caused by neuronal dysfunction without an apparent loss of neurons. The same single mutation (GAG deletion) causes most cases and results in loss of a glutamic acid (E) in the carboxy terminal region of torsinA (Delta302/303). To model the neuronal involvement, adult rat primary sensory dorsal root ganglion neurons in culture were infected with lentivirus vectors expressing human wild-type or mutant torsinA. Expression of the mutant protein resulted in formation of torsinA-positive perinuclear inclusions. When the cells were co-infected with lentivirus vectors expressing the mutant torsinA message and a shRNA selectively targeting this message, inclusion formation was blocked. Vector-delivered siRNAs have the potential to decrease the adverse effects of this mutant protein in neurons without affecting wild-type protein.


Assuntos
Chaperonas Moleculares/genética , Neurônios/ultraestrutura , Interferência de RNA/fisiologia , Animais , Células Cultivadas , Clonagem Molecular , Gânglios Espinais/citologia , Gânglios Espinais/ultraestrutura , Deleção de Genes , Genes gag/fisiologia , Vetores Genéticos , Ácido Glutâmico/metabolismo , Humanos , Lentivirus/genética , Oligonucleotídeos/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
13.
Mol Pain ; 1: 36, 2005 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-16354295

RESUMO

BACKGROUND: Pain is elicited by cold, and a major feature of many neuropathic pain states is that normally innocuous cool stimuli begin to produce pain (cold allodynia). To expand our understanding of cold induced pain states we have studied cold pain behaviors over a range of temperatures in several animal models of chronic pain. RESULTS: We demonstrate that a Peltier-cooled cold plate with +/- 1 degrees C sensitivity enables quantitative measurement of a detection withdrawal response to cold stimuli in unrestrained rats. In naïve rats the threshold for eliciting cold pain behavior is 5 degrees C. The withdrawal threshold for cold allodynia is 15 degrees C in both the spared nerve injury and spinal nerve ligation models of neuropathic pain. Cold hyperalgesia is present in the spared nerve injury model animals, manifesting as a reduced latency of withdrawal response threshold at temperatures that elicit cold pain in naïve rats. We also show that following the peripheral inflammation produced by intraplantar injection of complete Freund's adjuvant, a hypersensitivity to cold occurs. CONCLUSION: The peltier-cooled provides an effective means of assaying cold sensitivity in unrestrained rats. Behavioral testing of cold allodynia, hyperalgesia and pain will greatly facilitate the study of the neurobiological mechanisms involved in cold/cool sensations and enable measurement of the efficacy of pharmacological treatments to reduce these symptoms.


Assuntos
Temperatura Baixa/efeitos adversos , Hiperalgesia/diagnóstico , Medição da Dor/métodos , Dor/diagnóstico , Nervos Periféricos/fisiopatologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Animais , Comportamento Animal/fisiologia , Denervação , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Inflamação/induzido quimicamente , Inflamação/complicações , Inflamação/fisiopatologia , Ligadura , Masculino , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Dor/induzido quimicamente , Dor/fisiopatologia , Medição da Dor/normas , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Traumatismos dos Nervos Periféricos , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiopatologia , Nervos Espinhais/lesões , Nervos Espinhais/fisiopatologia , Vigília/fisiologia
14.
Pain ; 117(1-2): 77-87, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16098668

RESUMO

In both the spared nerve injury (SNI) and spinal nerve ligation (SNL) rat peripheral neuropathic pain models the presynaptic inhibitory effect of the mu opioid receptor (MOR) agonist (DAMGO) on primary afferent-evoked excitatory postsynaptic currents (EPSCs) and miniature EPSCs in superficial dorsal horn neurons is substantially reduced, but only in those spinal cord segments innervated by injured primary afferents. The two nerve injury models also reduce the postsynaptic potassium channel opening action of DAMGO on lamina II spinal cord neurons, but again only in segments receiving injured afferent input. The inhibitory action of DAMGO on ERK (extracellular signal-regulated kinase) activation in dorsal horn neurons is also reduced in affected segments following nerve injury. MOR expression decreases substantially in injured dorsal root ganglion neurons (DRG), while intact neighboring DRGs are unaffected. Decreased activation of MOR on injured primary afferent central terminals and the second order neurons they innervate may minimize any reduction by opioids of the spontaneous pain mediated by ectopic input from axotomized small diameter afferents. Retention of MOR expression and activity in nearby non-injured afferents will enable, however, an opioid-mediated reduction of stimulus-evoked and spontaneous pain carried by intact nociceptor afferents and we find that intrathecal DAMGO (1000 ng) reduces mechanical hypersensitivity in rats with SNL. Axotomy-induced changes in MOR may contribute to opioid- insensitive components of neuropathic pain while the absence of these changes in intact afferents may contribute to the opioid sensitive components.


Assuntos
Neurônios/fisiologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/fisiopatologia , Receptores Opioides mu/metabolismo , Medula Espinal , Sinapses/fisiologia , Analgésicos Opioides/farmacologia , Animais , Northern Blotting/métodos , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Ativação Enzimática , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lateralidade Funcional , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Neurofilamentos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Medição da Dor/métodos , Limiar da Dor/fisiologia , Técnicas de Patch-Clamp/métodos , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Estimulação Física/métodos , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Sinapses/efeitos dos fármacos , Sinapses/patologia
15.
Pain ; 100(1-2): 155-62, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12435468

RESUMO

Neuropathic pain syndromes are characterized by spontaneous pain and by stimulus-evoked allodynia and hyperalgesia. Stimulus-induced pain, i.e. the capacity of external stimuli to alter sensory processing so as to generate a pain hypersensitivity that outlasts the initiating stimulus, is usually present only after intense activation of nociceptors. In abnormal pain states, however, such as after capsaicin injection or inflammation, a stimulus-induced incremental pain can be generated by repetitive light touch, termed progressive tactile hypersensitivity (PTH). In the present study, we have examined whether PTH also occurs in two experimental models of neuropathic pain: a crush injury of the sciatic nerve and the spared nerve injury (SNI) model. When applied during the first weeks after injury to the territory of the injured crushed nerve, repeated low-intensity mechanical stimulation did not change the mechanical withdrawal threshold response. However, 10 weeks and after, the same repeated stimulation induced a progressive tactile hypersensitivity that persisted after discontinuation of the tactile stimulation. Following SNI, repeated stimulation of the hypersensitive skin territory, corresponding to the intact spared sural nerve, never induced PTH. Tactile stimulation of regenerating afferents but not spared non-injured afferents, can induce, therefore, PTH and such a stimulus-induced alteration in pain processing may contribute to clinical neuropathic pain.


Assuntos
Hiperalgesia/fisiopatologia , Ciática/fisiopatologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Masculino , Compressão Nervosa , Estimulação Física , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/lesões , Nervo Isquiático/fisiopatologia
16.
Pain ; 77(1): 49-57, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9755018

RESUMO

Normally-innocuous low-intensity tactile stimuli applied to inflamed tissue induce a progressive decrease in the mechanical flexion withdrawal threshold, the phenomenon of progressive tactile hypersensitivity (PTH). The effects of the mu opioid receptor agonist morphine, the non-competitive NMDA receptor antagonist MK801 and the tachykinin NK1 receptor antagonist RP67580 on the development and maintenance of PTH has now been investigated behaviourally in rats inflamed 48 h earlier by intraplantar complete Freund's adjuvant injection. A standard protocol of eight light tactile stimuli applied to the dorsum of the inflamed paw every 4 s at 5 min intervals resulted, over 60 min, in a 70% fall in mechanical threshold from the pre-conditioning baseline value. Morphine administered before the tactile stimuli at 0.05 mg/kg i.p. had no effect on either baseline thresholds or PTH. At 0.5 mg/kg, morphine prevented the establishment of PTH without changing baseline thresholds. At 5 mg/kg morphine produced analgesia, increasing thresholds above the baseline. MK801 pre-treatment at 0.01 and 0.001 mg/kg i.p. significantly attenuated the development of progressive tactile hyperalgesia without an effect on basal thresholds. RP67580 pre-treatment at 0.1 mg/kg i.p. had no effect, but at both I and 10 mg/kg, attenuated progressive tactile hypersensitivity without changing baseline values. To test the effect of the drugs on established PTH, they were administered 90 min after the commencement of intermittent tactile stimulation to the inflamed hindpaw, when thresholds had reached a plateau. Morphine (0.5 mg/kg) and MK801 (0.01 mg/kg) produced only a small reduction in sensitivity and RP67580 (1 mg/kg) had no effect. These results suggest that the induction of inflammatory progressive tactile hypersensitivity is sensitive to morphine, and to a lesser extent NMDA and NKI receptor antagonists, but these compounds at a dose that do not alter baseline values, do not normalise established tactile hypersensitivity.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos/farmacologia , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hiperalgesia/fisiopatologia , Indóis/farmacologia , Inflamação/complicações , Morfina/farmacologia , Tato/fisiologia , Animais , Membro Posterior , Hiperalgesia/etiologia , Isoindóis , Masculino , Estimulação Física , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de Taquicininas/antagonistas & inibidores
17.
BMC Neurosci ; 3: 16, 2002 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-12401135

RESUMO

BACKGROUND: Rat oligonucleotide microarrays were used to detect changes in gene expression in the dorsal root ganglion (DRG) 3 days following sciatic nerve transection (axotomy). Two comparisons were made using two sets of triplicate microarrays, naïve versus naïve and naïve versus axotomy. RESULTS: Microarray variability was assessed using the naïve versus naïve comparison. These results support use of a P < 0.05 significance threshold for detecting regulated genes, despite the large number of hypothesis tests required. For the naïve versus axotomy comparison, a 2-fold cut off alone led to an estimated error rate of 16%; combining a >1.5-fold expression change and P < 0.05 significance reduced the estimated error to 5%. The 2-fold cut off identified 178 genes while the combined >1.5-fold and P < 0.05 criteria generated 240 putatively regulated genes, which we have listed. Many of these have not been described as regulated in the DRG by axotomy. Northern blot, quantitative slot blots and in situ hybridization verified the expression of 24 transcripts. These data showed an 83% concordance rate with the arrays; most mismatches represent genes with low expression levels reflecting limits of array sensitivity. A significant correlation was found between actual mRNA differences and relative changes between microarrays (r2 = 0.8567). Temporal patterns of individual genes regulation varied. CONCLUSIONS: We identify parameters for microarray analysis which reduce error while identifying many putatively regulated genes. Functional classification of these genes suggest reorganization of cell structural components, activation of genes expressed by immune and inflammatory cells and down-regulation of genes involved in neurotransmission.


Assuntos
Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Traumatismos dos Nervos Periféricos , Neuropatia Ciática/fisiopatologia , Animais , Apoptose/genética , Axotomia , Northern Blotting , Citoesqueleto/genética , Modelos Animais de Doenças , Gânglios Espinais/citologia , Genoma , Hibridização In Situ , Masculino , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Nervos Periféricos/fisiopatologia , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
18.
Neuropharmacology ; 79: 136-51, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24269608

RESUMO

Effective relief from chronic hypersensitive pain states remains an unmet need. Here we report the discovery that the TRPM8 ion channel, co-operating with the 5-HT(1B) receptor (5-HT(1B)R) in a subset of sensory afferents, exerts an influence at the spinal cord level to suppress central hypersensitivity in pain processing throughout the central nervous system. Using cell line models, ex vivo rat neural tissue and in vivo pain models, we assessed functional Ca(2+) fluorometric responses, protein:protein interactions, immuno-localisation and reflex pain behaviours, with pharmacological and molecular interventions. We report 5-HT(1B)R expression in many TRPM8-containing afferents and direct interaction of these proteins in a novel multi-protein signalling complex, which includes phospholipase D1 (PLD1). We provide evidence that the 5-HT(1B)R activates PLD1 to subsequently activate PIP 5-kinase and generate PIP2, an allosteric enhancer of TRPM8, achieving a several-fold increase in potency of TRPM8 activation. The enhanced activation responses of synaptoneurosomes prepared from spinal cord and cortical regions of animals with a chronic inflammatory pain state are inhibited by TRPM8 activators that were applied in vivo topically to the skin, an effect potentiated by co-administered 5-HT(1B)R agonists and attenuated by 5-HT(1B)R antagonists, while 5-HT(1B)R agents alone had no detectable effect. Corresponding results are seen when assessing reflex behaviours in inflammatory and neuropathic pain models. Control experiments with alternative receptor/TRP channel combinations reveal no such synergy. Identification of this novel receptor/effector/channel complex and its impact on nociceptive processing give new insights into possible strategies for enhanced analgesia in chronic pain.


Assuntos
Dor/metabolismo , Fosfolipase D/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiopatologia , Células HEK293 , Humanos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/fisiopatologia , Dor/tratamento farmacológico , Ratos , Ratos Wistar , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Antagonistas do Receptor 5-HT1 de Serotonina/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiopatologia , Canal de Cátion TRPA1 , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo
19.
Neurosci Res ; 74(3-4): 230-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23131427

RESUMO

Many clinical cases of chronic pain exhibit both neuropathic and inflammatory components. In contrast, most animal models of chronic pain focus on one type of injury alone. Here we present a novel combined model of both neuropathic and inflammatory pain and characterise its distinctive properties. This combined model of chronic constriction injury (CCI) and intraplantar Complete Freund's Adjuvant (CFA) injection results in enhanced mechanical allodynia, thermal hyperalgesia, a static weight bearing deficit, and notably pronounced spontaneous foot lifting (SFL) behaviour (which under our conditions was not seen in either individual model and may reflect ongoing/spontaneous pain). Dorsal root ganglion (DRG) expression of Activating Transcription Factor-3 (ATF-3), a marker of axonal injury, was no greater in the combined model than CCI alone. Initial pharmacological characterisation of the new model showed that the SFL was reversed by gabapentin or diclofenac, typical analgesics for neuropathic or inflammatory pain respectively, but not by mexiletine, a Na(+) channel blocker effective in both neuropathic and inflammatory pain models. Static weight bearing deficit was moderately reduced by gabapentin, whereas only diclofenac reversed mechanical allodynia. This novel animal model of chronic pain may prove a useful test-bed for further analysing the pharmacological susceptibility of complicated clinical pain states.


Assuntos
Dor Crônica , Modelos Animais de Doenças , Inflamação , Neuralgia , Fator 3 Ativador da Transcrição/análise , Fator 3 Ativador da Transcrição/biossíntese , Adjuvantes Imunológicos/toxicidade , Animais , Comportamento Animal/fisiologia , Dor Crônica/metabolismo , Dor Crônica/fisiopatologia , Adjuvante de Freund/toxicidade , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/fisiopatologia , Masculino , Neuralgia/metabolismo , Neuralgia/fisiopatologia , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/lesões
20.
Mol Cell Neurosci ; 36(2): 185-94, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17702601

RESUMO

Axonal regeneration within the CNS fails due to the growth inhibitory environment and the limited intrinsic growth capacity of injured neurons. Injury to DRG peripheral axons induces expression of growth associated genes including members of the glial-derived neurotrophic factor (GDNF) signaling pathway and "preconditions" the injured cells into an active growth state, enhancing growth of their centrally projecting axons. Here, we show that preconditioning DRG neurons prior to culturing increased neurite outgrowth, which was further enhanced by GDNF in a bell-shaped growth response curve. In vivo, GDNF delivered directly to DRG cell bodies facilitated the preconditioning effect, further enhancing axonal regeneration beyond spinal cord lesions. Consistent with the in vitro results, the in vivo effect was seen only at low GDNF concentrations. We conclude that peripheral nerve injury upregulates GDNF signaling pathway components and that exogenous GDNF treatment selectively promotes axonal growth of injury-primed sensory neurons in a concentration-dependent fashion.


Assuntos
Gânglios Espinais/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Traumatismos da Medula Espinal/patologia , Animais , Relação Dose-Resposta a Droga , Masculino , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA