Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
2.
J Dent Res ; 95(10): 1084-92, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27339423

RESUMO

In 2006, the OPPERA project (Orofacial Pain: Prospective Evaluation and Risk Assessment) set out to identify risk factors for development of painful temporomandibular disorder (TMD). A decade later, this review summarizes its key findings. At 4 US study sites, OPPERA recruited and examined 3,258 community-based TMD-free adults assessing genetic and phenotypic measures of biological, psychosocial, clinical, and health status characteristics. During follow-up, 4% of participants per annum developed clinically verified TMD, although that was a "symptom iceberg" when compared with the 19% annual rate of facial pain symptoms. The most influential predictors of clinical TMD were simple checklists of comorbid health conditions and nonpainful orofacial symptoms. Self-reports of jaw parafunction were markedly stronger predictors than corresponding examiner assessments. The strongest psychosocial predictor was frequency of somatic symptoms, although not somatic reactivity. Pressure pain thresholds measured at cranial sites only weakly predicted incident TMD yet were strongly associated with chronic TMD, cross-sectionally, in OPPERA's separate case-control study. The puzzle was resolved in OPPERA's nested case-control study where repeated measures of pressure pain thresholds revealed fluctuation that coincided with TMD's onset, persistence, and recovery but did not predict its incidence. The nested case-control study likewise furnished novel evidence that deteriorating sleep quality predicted TMD incidence. Three hundred genes were investigated, implicating 6 single-nucleotide polymorphisms (SNPs) as risk factors for chronic TMD, while another 6 SNPs were associated with intermediate phenotypes for TMD. One study identified a serotonergic pathway in which multiple SNPs influenced risk of chronic TMD. Two other studies investigating gene-environment interactions found that effects of stress on pain were modified by variation in the gene encoding catechol O-methyltransferase. Lessons learned from OPPERA have verified some implicated risk factors for TMD and refuted others, redirecting our thinking. Now it is time to apply those lessons to studies investigating treatment and prevention of TMD.


Assuntos
Dor Facial/genética , Dor Facial/fisiopatologia , Transtornos da Articulação Temporomandibular/genética , Transtornos da Articulação Temporomandibular/fisiopatologia , Adulto , Estudos Transversais , Interação Gene-Ambiente , Genótipo , Humanos , Medição da Dor , Fenótipo , Polimorfismo de Nucleotídeo Único , Medição de Risco , Fatores de Risco , Estados Unidos
3.
Osteoarthritis Cartilage ; 21(9): 1308-15, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23973145

RESUMO

OBJECTIVE: Osteoarthritis (OA) is a complex and painful disease of the whole joint. At present there are no satisfying agents for treating OA. To promote OA research and improved treatment, this review summarizes current preclinical evidence on the development of OA. METHODS: Preclinical OA research was searched and key findings are summarized and commented. RESULTS: Mechanisms of OA-associated pain have been studied in rodent knee OA models produced by intra-knee injection of the chondrocyte glycolytic inhibitor mono-iodoacetate (MIA), surgery, or spontaneous development in some species. These models are clinically relevant in terms of histological damage and functional changes, and are used to study mechanisms underlying mechanical, thermal, ambulatory, body weight supporting-evoked, and ongoing OA pain. Recent peripheral, spinal, and supraspinal biochemical and electrophysiological studies in these models suggest that peripheral pro-inflammatory mediators and neuropeptides sensitize knee nociceptors. Spinal cytokines and neuropeptides promote OA pain, and peripheral and spinal cannabinoids inhibit OA pain respectively through cannabinoid-1 (CB1) and CB1/CB2 receptors. TRPV1 and metalloproteinases contribute and supraspinal descending facilitation of 5-hydroxytryptamine (5-HT)/5-HT 3 receptors may also contribute to OA pain. Conditioned place preference tests demonstrate that OA pain induces aversive behaviors, suggesting the involvement of brain. During OA, brain functional connectivity is enhanced, but at present it is unclear how this change is related to OA pain. CONCLUSION: Animal studies demonstrate that peripheral and central sensitization contributes to OA pain, involving inflammatory cytokines, neuropeptides, and a variety of chemical mediators. Interestingly, brainstem descending facilitation of 5-HT/5-HT3 receptors plays a role OA pain.


Assuntos
Artralgia/fisiopatologia , Artrite Experimental/fisiopatologia , Modelos Animais de Doenças , Hiperalgesia/fisiopatologia , Osteoartrite do Joelho/fisiopatologia , Analgésicos/farmacologia , Animais , Artralgia/tratamento farmacológico , Artrite Experimental/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Osteoartrite do Joelho/tratamento farmacológico
4.
J Dent Res ; 92(7 Suppl): 70S-7S, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23690360

RESUMO

The authors tested the hypothesis that obstructive sleep apnea (OSA) signs/symptoms are associated with the occurrence of temporomandibular disorder (TMD), using the OPPERA prospective cohort study of adults aged 18 to 44 years at enrollment (n = 2,604) and the OPPERA case-control study of chronic TMD (n = 1,716). In both the OPPERA cohort and case-control studies, TMD was examiner determined according to established research diagnostic criteria. People were considered to have high likelihood of OSA if they reported a history of sleep apnea or ≥ 2 hallmarks of OSA: loud snoring, daytime sleepiness, witnessed apnea, and hypertension. Cox proportional hazards regression estimated hazard ratios (HRs) and 95% confidence limits (CL) for first-onset TMD. Logistic regression estimated odds ratios (OR) and 95% CL for chronic TMD. In the cohort, 248 individuals developed first-onset TMD during the median 2.8-year follow-up. High likelihood of OSA was associated with greater incidence of first-onset TMD (adjusted HR = 1.73; 95% CL, 1.14, 2.62). In the case-control study, high likelihood of OSA was associated with higher odds of chronic TMD (adjusted OR = 3.63; 95% CL, 2.03, 6.52). Both studies supported a significant association of OSA symptoms and TMD, with prospective cohort evidence finding that OSA symptoms preceded first-onset TMD.


Assuntos
Apneia Obstrutiva do Sono/complicações , Transtornos da Articulação Temporomandibular/complicações , Adolescente , Adulto , Negro ou Afro-Americano , Fatores Etários , Pressão Sanguínea/fisiologia , Índice de Massa Corporal , Estudos de Casos e Controles , Estudos de Coortes , Eletrocardiografia , Feminino , Seguimentos , Frequência Cardíaca/fisiologia , Humanos , Hipertensão/complicações , Masculino , Obesidade/complicações , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores Sexuais , Fases do Sono/fisiologia , Fumar , Ronco/complicações , População Branca , Adulto Jovem
5.
Open Pain J ; 2010(3): 144-152, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21170295

RESUMO

We tested the hypothesis that primary afferent inputs play a role in astroglial hyperactivity after tissue injury. We first injected complete Freund's adjuvant (CFA, 0.05 ml, 1:1 oil/saline) into the masseter muscle, which upregulated glial fibrillary acidic protein (GFAP), a marker of astrocytes, interleukin (IL)-1ß an inflammatory cytokine, and phosphorylation of serine896 of the NR1 subunit (P-NR1) of the NMDA receptor in the subnuclei interpolaris/caudalis (Vi/Vc) transition zone, an important structure for processing trigeminal nociceptive input. Local anesthetic block with lidocaine (2%) of the masseter muscle at 10 min prior to injection of CFA into the same site significantly reduced the CFA-induced increase in GFAP, IL-1ß and P-NR1 (p<0.05, n=4/group). We then tested the effect of peripheral electrical stimulation (ES). The ES protocol was burst stimulation consisting of trains of 4 square pulses (10-100 Hz, 0.1-3 mA, 0.5 ms pulse width). Under pentobarbital anesthesia, an ES was delivered every 0.2 s for a total of 30 min. The Vi/Vc tissues were processed for immunohistochemistry or western blot analysis at 10-120 min after ES. Compared to naive and SHAM-treated rats, there was increased immunoreactivity against GFAP, IL-1ß and P-NR1 in the Vi/Vc in rats receiving ES. Double staining showed that IL-1ß was selectively localized in GFAP-positive astroglia, and P-NR1-immunoreactivity was localized to neurons. These findings indicate that primary afferent inputs are necessary and sufficient to induce astroglial hyperactivity and upregulation of IL-1ß, as well as neuronal NMDA receptor phosphorylation.

6.
Neuroscience ; 171(4): 1341-56, 2010 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20888891

RESUMO

Hyperalgesia in animal injury models is linked to activation of descending raphespinal modulatory circuits originating in the rostral ventromedial medulla (RVM). A neurokinin-1 (NK-1) receptor antagonist microinjected into the RVM before or after inflammation produced by complete Freund's adjuvant (CFA) resulted in an attenuation of thermal hyperalgesia. A transient (acute) or a continuous infusion of Substance P (SP) microinjected into the RVM of non-inflamed animals led to similar pain hypersensitivity. Intrathecal pretreatment or post-treatment of a 5-HT3 receptor antagonist (Y-25130 or ondansetron) blocked the SP-induced hyperalgesia. The SP-induced hyperalgesia was both GABA(A) and NMDA receptor-dependent after pre- and post-treatment with selective antagonists at the spinal level. A microinjection of SP into the RVM also led to increased NMDA NR1 receptor subunit phosphorylation in spinal cord tissue. The GABA(A) receptor-mediated hyperalgesia involved a shift in the anionic gradient in dorsal horn nociceptive neurons and an increase in phosphorylated NKCC1 protein (isoform of the Na-K-Cl cotransporter). Following a low dose of SP infused into the RVM, intrathecal muscimol (GABA(A) agonist) increased SP-induced thermal hyperalgesia, phosphorylated NKCC1 protein expression, and NMDA NR1 subunit phosphorylation in the spinal cord. The thermal hyperalgesia was blocked by intrathecal gabazine, the GABA(A) receptor antagonist, and MK-801, the NMDA receptor channel blocker. These findings indicate that NK-1 receptors in the RVM are involved in SP-induced thermal hyperalgesia, this hyperalgesia is 5-HT3-receptor dependent at the spinal level, and involves the functional interaction of spinal GABA(A) and NMDA receptors.


Assuntos
Hiperalgesia/patologia , Bulbo/metabolismo , Limiar da Dor/fisiologia , Receptores da Neurocinina-1/metabolismo , Medula Espinal/metabolismo , Animais , Comportamento Animal , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Adjuvante de Freund , GABAérgicos/farmacologia , Hiperalgesia/etiologia , Hiperalgesia/prevenção & controle , Técnicas In Vitro , Inflamação/induzido quimicamente , Inflamação/complicações , Masculino , Bulbo/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Microinjeções/métodos , Muscimol/farmacologia , Ondansetron/farmacologia , Oxazinas/farmacologia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/fisiologia , Piridazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Antagonistas da Serotonina/farmacologia , Medula Espinal/citologia , Substância P/farmacologia , Triptofano/análogos & derivados , Triptofano/farmacologia , Regulação para Cima/efeitos dos fármacos
7.
Open Pain J ; 2: 76-83, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20221418

RESUMO

Our recent studies indicate that the prototypic proinflammatory cytokine IL-1ß is upregulated in astroglial cells in the trigeminal interplolaris/caudalis (Vi/Vc) transition zone, a region of the spinal trigeminal complex involved in trigeminal pain processing, after masseter muscle inflammation. Here we investigated the effect of microinjection of IL-1ß into the Vi/Vc transition zone on orofacial nociception. The mechanical sensitivity of the orofacial site was assessed with von Frey microfilaments. The EF(50) values, defined as the von Frey filament force (g) that produces a 50% response frequency, were derived and used as a measure of mechanical sensitivity. A significant reduction in EF(50) indicates the occurrence of mechanical hyperalgesia/allodynia. Unilateral intra-Vi/Vc IL-1ß (0.016-160 fmol) produced hyperalgesia/allodynia dose-dependently, which appeared at bilateral facial sites. The hyperalgesia was detectable as early as 30 min and lasted for 2-6 h (n=6, p<0.01). Intra-Vi/Vc pretreatment with an IL-1receptor antagonist (1 nmol) attenuated the IL-1ß-induced hyperalgesia (p<0.01). Pre-injection of AP-5 (10 pmol) and MK-801 (20 pmol), two NMDA receptor antagonists, significantly attenuated IL-1ß-induced hyperalgesia (p<0.05). Pretreatment with glial inhibitors fluorocitrate (120 pmol), minocycline (200 pmol) and propentofylline (10 pmol) did not attenuate IL-1ß-induced hyperalgesia. Excitotoxic lesions of the rostral ventromedial medulla with ibotenic acid (2 µg) abolished IL-1ß-induced contralateral hyperalgesia, suggesting a contribution of descending facilitatory drive. These results suggest that the IL-1ß-produced effect on nociception was downstream to glial activation and involves interaction with NMDA receptors.

8.
Neuroscience ; 133(1): 231-43, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15893646

RESUMO

Previous studies demonstrated that nursing or intraoral infusion of certain components of mother's milk (e.g. sugars and fats) produces calming and opiate receptor-dependent analgesia in newborn rats and humans. However, the neural circuitry underlying such analgesia is unknown. The aim of the present study was to specify the central pathways by which taste stimuli engage neural antinociceptive mechanisms. For this purpose, midcollicular transactions were used to investigate the role of the forebrain in analgesia elicited by intraoral infusion of 0.2 M sucrose in neonatal rats. Sucrose-induced analgesia persisted, and was enhanced, following midcollicular transection, indicating that it did not require neural circuits confined to the forebrain. Fos immunohistochemistry was used to identify brainstem neurons activated by a brief (90 s) intraoral infusion of a small volume (90 microl, 0.2M) of sucrose or a salt solution (0.1 M ammonium chloride) in 10-day-old rat pups. Compared with control groups (intact, cannula, distilled water), both sucrose and ammonium chloride induced Fos expression in the rostral nucleus tractus solitarius, the first relay in the ascending gustatory pathway. Sucrose also elicited Fos expression in several brainstem areas associated with centrally mediated analgesia, including the periaqueductal gray and the nucleus raphe magnus. Taken together, these findings demonstrate that analgesia elicited by intraoral sucrose does not require involvement of the forebrain. Intraoral sucrose activates neurons in the periaqueductal gray and nucleus raphe magnus, two key brainstem sites critically involved in descending pain modulation.


Assuntos
Analgesia , Analgésicos , Tronco Encefálico/fisiologia , Sacarose/farmacologia , Administração Oral , Animais , Animais Recém-Nascidos , Feminino , Imuno-Histoquímica , Masculino , Vias Neurais/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Ratos , Ratos Sprague-Dawley , Sacarose/administração & dosagem , Colículos Superiores/fisiologia
9.
Pain ; 110(3): 588-596, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15288399

RESUMO

Recently, several studies have suggested that neonatal noxious insult could alter future responses to painful stimuli. However, the manifestations, mechanisms, and even developmental nature of these alterations remain a matter of controversy. In part, this is due to the lack of detailed information on the neonatal sensitive period(s) during which noxious stimulation influences future nociception, and the time-course and distribution of the resultant abnormalities. The present paper describes these parameters in a rat model of short-lasting ( approximately 24 h) neonatal local inflammation of a hindpaw produced by injection of 0.25% carrageenan (1 microl/g). Examinations of paw withdrawal responses to thermal and mechanical stimulations in adult animals, which as neonates were subjected to this insult, showed that the previously-reported long-term hypoalgesia and hyperalgesia are not mutually exclusive outcomes of early noxious experience. Long-term hypoalgesia was apparent at the basal conditions and was equally strong in the previously injured and uninjured paws, which suggests a globally-driven deficit. In contrast, long-term excessive hyperalgesia had the strongest manifestation in the neonatally-injured paw after re-inflammation, indicating significant segmental involvement in its generation. The differences between mechanisms underlying the observed hypoalgesia and hyperalgesia are further underscored by the finding that, while the former is detectable only after animals reach the second month of life, the latter is elicitable immediately upon cessation of the initial neonatal inflammation. Nevertheless, we detected a significant overlap in the neonatal sensitive periods for generation of these effects (both occurring within the first postnatal week). Also, neither the basal hypoalgesia nor excessive re-inflammation-associated hyperalgesia subsided with age and were detectable in 120-125-day-old rats. These finding provide a framework within which the entire complex of long-term effects of early noxious experience can be understood and examined.


Assuntos
Inflamação/fisiopatologia , Medição da Dor/métodos , Dor/patologia , Dor/fisiopatologia , Animais , Animais Recém-Nascidos , Carragenina/toxicidade , Feminino , Inflamação/induzido quimicamente , Masculino , Dor/induzido quimicamente , Gravidez , Ratos , Ratos Sprague-Dawley , Tempo , Fatores de Tempo
10.
Pain ; 109(3): 507-513, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15157713

RESUMO

Significant analgesic and calming effects in human infants and neonatal rodents are produced by orogustatory and orotactile stimuli associated with nursing. These naturally occurring analgesic stimuli may help to protect the vulnerable developing nervous system from the long-term effects of neonatal tissue injury. However, the efficacy of orotactile-induced analgesia across the pre-weaning period, as well as its effects on persistent inflammatory pain, is unknown. Here, we investigated the developmental profile of analgesia produced by orotactile stimulation during non-nutritive suckling in rats. The effects of suckling, as compared to non-suckling littermates, on nocifensive withdrawal responses to thermal and mechanical stimuli were examined at postnatal (P) days P0, P3, P10, P17 and P21. In some rats, Complete Freund's adjuvant (CFA) was injected in a fore- or hindpaw to produce inflammation. For thermal stimuli, suckling significantly increased forepaw withdrawal latencies at P3, P10 and P17, while hindpaw responses were increased at P3 and P10, but not at P17. In inflamed pups, suckling increased fore- and hindpaw response latencies at P10 and P17, but not at P0 or P21. Suckling-induced analgesia was naloxone-insensitive. For mechanical stimuli, suckling-induced analgesia was present at P3, P10 and P17, but not at P21, for both fore- and hindpaws in naïve and inflamed animals. Additionally, suckling had a small but significant effect at P0 for the forepaw in inflamed pups. In nearly all experiments, the peak effect of suckling for thermal and mechanical stimuli occurred at P10. These results indicate that orotactile analgesia, like orogustatory analgesia, is absent or minimal at P0, appears consistently at approximately P3 and is maximal at P10. Unlike gustatory analgesia in rats however, orotactile analgesia persists at least to P17. Orotactile stimulation during suckling effectively reduces transient pain elicited by thermal and mechanical stimuli, as well as persistent hyperalgesia and allodynia caused by inflammation.


Assuntos
Analgesia , Animais Lactentes/fisiologia , Inibição Neural/fisiologia , Manejo da Dor , Dor/fisiopatologia , Comportamento de Sucção/fisiologia , Doença Aguda , Vias Aferentes/crescimento & desenvolvimento , Vias Aferentes/fisiologia , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/fisiologia , Doença Crônica , Modelos Animais de Doenças , Feminino , Hiperalgesia/fisiopatologia , Hiperalgesia/terapia , Inflamação/fisiopatologia , Inflamação/terapia , Masculino , Mecanorreceptores/fisiologia , Boca/inervação , Boca/fisiologia , Medição da Dor , Estimulação Física , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Reflexo/fisiologia , Pele/inervação , Pele/fisiopatologia , Tato/fisiologia
11.
Pain ; 97(1-2): 171-81, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12031790

RESUMO

Previous studies indicate that descending modulation of nociception is progressively increased following persistent inflammation. The present study was designed to further examine the role of supraspinal neurons in descending modulation following persistent inflammation. Constant levels of paw withdrawal (PW) and tail flick (TF) latencies to noxious heat stimuli were achieved in lightly anesthetized rats (pentobarbital sodium 3-10 mg/kg/h, i.v.). Electrical stimulation (ES, 0.1 ms, 100 Hz, 20-200 A) was delivered to the rostral ventromedial medulla (RVM), mainly the nucleus raphe magnus (NRM). ES produced intensity-dependent inhibition of PW and TF. Following a unilateral hindpaw inflammation produced by injection of complete Freund's adjuvant (CFA), ES-produced inhibition underwent time-dependent changes. There was an initial decrease at 3 h after inflammation and a subsequent increase after inflammation in the excitability of RVM neurons and the inhibition of nocifensive responses. These changes were most robust after stimulation of the inflamed paw although similar findings were seen on the non-inflamed paw and tail. The inflammation-induced dynamic changes in descending modulation appeared to be correlated with changes in the activation of the N-methyl--aspartate (NMDA) excitatory amino acid receptor. Microinjection of an NMDA receptor antagonist, AP5 (1 pmol), resulted in an increase in the current intensity required for inhibition of the PW and TF. The effect of AP5 was less at 3 h after inflammation and significantly greater at 11-24 h after inflammation. In a subsequent experiment, ES-produced inhibition of nocifensive responses after inflammation was examined following selective chemical lesions of the nuclei reticularis gigantocellularis (NGC). Compared to vehicle-injected animals, microinjection of a soma-selective excitotoxin, ibotenic acid, enhanced ES-produced inhibition at 3 h but not at 24 h after inflammation. We propose that these time course changes reflect dynamic alterations in concomitant descending facilitation and inhibition. At early time points, NMDA receptor and NGC activation enhance descending facilitation; as time progresses, the dose-response curve of NMDA shifts to the left and descending inhibition dominates and masks any descending facilitation.


Assuntos
Hiperalgesia/fisiopatologia , Bulbo/fisiologia , Inibição Neural/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Anestesia , Animais , Estimulação Elétrica , Agonistas de Aminoácidos Excitatórios , Antagonistas de Aminoácidos Excitatórios/farmacologia , Temperatura Alta , Ácido Ibotênico , Masculino , Inflamação Neurogênica/fisiopatologia , Nociceptores/fisiologia , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação
12.
J Neurophysiol ; 87(2): 750-60, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11826044

RESUMO

Recent studies indicate that descending pain modulatory pathways undergo time-dependent changes in excitability following inflammation involving both facilitation and inhibition. The cellular and molecular mechanisms of these phenomena are unclear. In the present study, we examined N-methyl-D-aspartate (NMDA) receptor gene expression and neuronal activity in the rostral ventromedial medulla (RVM), a pivotal structure in pain modulatory circuitry, after complete Freund's adjuvant (CFA)-induced hindpaw inflammation. The reverse transcription polymerase chain reaction analysis indicated that there was an upregulation of mRNAs encoding NMDA receptor subunits in the RVM after inflammation. The increase in the NR1, NR2A, and NR2B receptor mRNAs started at 5 h, maintained for 1-7 days (P < 0.05-0.001) and returned to the control level at 14 days after inflammation. Western blot analysis indicated that the protein translation products of the NR2A subunit were also increased (P < 0.01). In single-unit extracellular recordings, we correlated RVM neuronal activity with the paw withdrawal response in rats with inflammation. We describe these RVM cells as on-, off-, and neutral-like cells because of their similarity to previous studies in which neuronal responses were correlated with tail-flick nocifensive behavior in the absence of inflammation. In contrast to previous studies in the absence of inflammation, using tail flick as a behavioral correlate, fewer off-like cells in naïve animals exhibited a complete pause before the paw withdrawal to a noxious thermal stimulus. The percentage of cells showing a pause of activity after noxious stimulation was further reduced after inflammation (chi(2) P < 0.0001 vs. naïve rats). Continuous neuronal recordings (3-6.5 h) revealed a phenotypic switch of RVM neurons during the development of inflammation: 11/15 neutral-like cells initially unresponsive to noxious stimuli exhibited and maintained response profiles characteristic of pain modulatory neurons (became off-like: n = 5; became on-like: n = 6). Neutral-like cells recorded in noninflamed animals did not show response profile changes during continuous recordings (5-5.5 h, n = 7). A population study (n = 165) confirmed an increase in on- and off-like cells and a decrease in neutral-like cells at 24 h after inflammation as compared with naïve rats (P < 0.001). These results suggest that enhanced NMDA receptor activation mediates time-dependent changes in excitability of RVM pain modulatory circuitry. The functional phenotypic switch of RVM neurons provides a novel mechanism underlying activity-dependent plasticity and enhanced net descending inhibition after inflammation.


Assuntos
Bulbo/citologia , Bulbo/fisiologia , Neurônios/fisiologia , Dor/fisiopatologia , Receptores de N-Metil-D-Aspartato/genética , Potenciais de Ação/fisiologia , Animais , Eletrofisiologia , Expressão Gênica/fisiologia , Inflamação/fisiopatologia , Masculino , Vias Neurais , Plasticidade Neuronal/fisiologia , Dor/imunologia , Fenótipo , Ratos , Ratos Sprague-Dawley
13.
Cells Tissues Organs ; 169(3): 238-47, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11455119

RESUMO

A rat model has been developed to characterize the responses of brainstem trigeminal neurons to orofacial deep and cutaneous tissue inflammation and hyperalgesia. Complete Freund's adjuvant (CFA) was injected unilaterally into the rat temporomandibular joint (TMJ) or perioral (PO) skin to produce inflammation in deep or cutaneous tissues, respectively. The TMJ and PO inflammation resulted in orofacial behavioral hyperalgesia and allodynia that peaked within 4-24 h and persisted for at least 2 weeks. Compared to cutaneous CFA injection, the injection of CFA into the TMJ produced a significantly stronger inflammation associated with a selective upregulation of preprodynorphin mRNA in the trigeminal spinal complex, an enhanced medullary dorsal horn hyperexcitability, and a greater trigeminal Fos protein expression, a marker of neuronal activation. The Fos-LI induced by TMJ inflammation persisted longer, was more intense, particularly in the superficial laminae, and more widespread rostrocaudally. Thus, the inflammatory irritant produces a stronger effect in deep than in cutaneous orofacial tissue. As there is heavy innervation of the TMJ by unmyelinated nerve endings, a strong nociceptive primary afferent barrage is expected following inflammation. An increase in TMJ C-fiber input after inflammation and strong central neuronal activation may initiate central hyperexcitability and contribute to persistent pain associated with temporomandibular disorders. Since deep inputs may be more effective in inducing central neuronal excitation than cutaneous inputs, greater sensory disturbances may occur in pain conditions involving deep tissues than in those involving cutaneous tissues.


Assuntos
Inflamação/fisiopatologia , Transtornos da Articulação Temporomandibular/fisiopatologia , Nervo Trigêmeo/fisiopatologia , Animais , Comportamento Animal , Dinorfinas/genética , Dor Facial/fisiopatologia , Inflamação/genética , Precursores de Proteínas/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Pele/fisiopatologia , Transtornos da Articulação Temporomandibular/etiologia , Transtornos da Articulação Temporomandibular/genética , Regulação para Cima
14.
Brain Res Mol Brain Res ; 88(1-2): 186-93, 2001 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-11295247

RESUMO

Glutamate receptors are involved in spinal nociceptive transmission and the development of persistent inflammatory hyperalgesia. It is unclear, however, whether there are changes in glutamate receptor gene expression associated with tissue injury. In the present study, we used reverse transcription-polymerase chain reaction (RT-PCR) to examine the modulation of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor gene expression in the rat spinal cord by inflammation. Inflammation was introduced into the hindpaw by intraplantar injection of 0.2 ml of complete Freund's adjuvant (CFA). At 2 h-14 d after inflammation, total RNAs from L4,5 spinal cord were used for RT-PCR with primers targeted at eight flip-flop splice variants of the AMPA receptor subunits. It was found that the GluR1-flop mRNA was up-regulated at 2 h-5 h (P<0.05), down-regulated at 3 d (P=0.05), and returned to control levels at 7 d following inflammation. The GluR2-flip and GluR3-flop mRNAs were up-regulated at 5 h-1 d (P<0.05) and returned to control levels at 3 d after inflammation. The GluR1-flip mRNA was not detected in the samples and the mRNAs for other splice variants did not exhibit significant changes. Immunocytochemical analysis of GluR1 and GluR2 subunits indicate that the protein translation products of these subunits were also increased in the spinal cord. These results demonstrate an increased expression of AMPA receptor subunits that correlates with the acute phase of CFA-induced inflammation and hyperalgesia. Selective changes in the expression of the flip-flop splice variants of the AMPA receptor suggest a reorganization of the composition of the AMPA receptor complex and its involvement in the development of inflammatory hyperalgesia.


Assuntos
Processamento Alternativo/fisiologia , Hiperalgesia/fisiopatologia , Inflamação Neurogênica/fisiopatologia , Receptores de AMPA/genética , Medula Espinal/imunologia , Animais , Adjuvante de Freund , Expressão Gênica/imunologia , Membro Posterior , Hiperalgesia/induzido quimicamente , Imuno-Histoquímica , Masculino , Inflamação Neurogênica/induzido quimicamente , Nociceptores/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/química
15.
Neuroreport ; 11(9): 1915-9, 2000 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-10884043

RESUMO

Brain stem descending pathways modulate spinal nociceptive transmission. In a lightly anesthetized rat preparation, we present evidence that such descending modulation undergoes time-dependent changes following persistent hindpaw inflammation. There was an initial decrease and a subsequent increase in the excitability of neurons in the rostral ventromedial medulla (RVM) involving facilitation and inhibition. These changes were most robust after stimulation of the inflamed paw although similar findings were seen on the non-inflamed paw and tail. The enhanced descending modulation appeared to be mediated by changes in the activation of the NMDA excitatory amino acid receptor. These findings demonstrate the dynamic plasticity of the pain modulating pathways in response to persistent tissue injury.


Assuntos
Inflamação/fisiopatologia , Bulbo/fisiopatologia , Plasticidade Neuronal , Dor/fisiopatologia , Animais , Comportamento Animal , Vias Eferentes/fisiopatologia , Estimulação Elétrica , Agonistas de Aminoácidos Excitatórios/farmacologia , Temperatura Alta , Masculino , N-Metilaspartato/farmacologia , Inibição Neural , Nociceptores/fisiopatologia , Medição da Dor , Estimulação Física , Ratos , Ratos Sprague-Dawley , Tempo de Reação , Receptores de Glutamato/fisiologia , Estimulação Química , Transmissão Sináptica/efeitos dos fármacos
16.
Brain Res ; 865(2): 272-7, 2000 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-10821931

RESUMO

The relationship between endogenous gonadal steroid levels and persistent or chronic pain is poorly understood. These studies used an inflammation model to examine the role of the gonadal steroid, progesterone, in the development of persistent pain and hyperalgesia in lactating ovary-intact and ovariectomized rats. The results indicate that constant high plasma levels of progesterone attenuate inflammatory hyperalgesia by a mechanism involving inhibition of N-methyl-D-aspartate receptor activation at the spinal cord level. Since the pattern of high progesterone in lactating rats mimics the progesterone component of the luteal phase of the human menstrual cycle, these findings have significance in persistent or chronic pain conditions that are most prevalent in females.


Assuntos
Hiperalgesia/sangue , Inflamação/sangue , Lactação/sangue , Progesterona/sangue , Animais , Estro/sangue , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Adjuvante de Freund , Hiperalgesia/induzido quimicamente , Inflamação/induzido quimicamente , N-Metilaspartato/farmacologia , Ovariectomia , Gravidez , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
17.
Eur J Pharmacol ; 387(2): 163-9, 2000 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-10650156

RESUMO

Inflammatory hyperalgesia was induced in wild-type, heterozygous and mu-opioid receptor knockout mice after an intraplantar injection of complete Freund's adjuvant. micro-Opioid receptor knockout mice exhibited faster recovery from hyperalgesia as compared to heterozygous (P<0.05) and wild-type (P<0.01) mice. Naloxone restored hyperalgesia in all genotypes. Naltrindole (delta-opioid receptor-selective antagonist) partially restored the hyperalgesia only in mu-opioid receptor knockout mice (P<0.001). Nor-binaltorphimine (kappa-opioid receptor-selective antagonist) had no effect. The mu-opioid receptor-selective agonist, [D-Ala(2), MePhe(4),Gly-ol(5)]enkephalin (DAMGO), reduced the hyperalgesia in heterozygous and wild-type but not in mu-opioid receptor knockout mice while U69,593 ¿(+)-(5alpha,7alpha, 8beta)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4. 5]dec-8-yl]-benzeneacetamide, kappa-opioid receptor-selective¿ produced similar effects in all mice. The delta-opioid receptor-selective agonists, [D-Pen(2), D-Pen(5)]enkephalin (DPDPE) and deltorphin ([D-Ala(2)]deltrophin-II), produced significantly greater antihyperalgesia in knockout mice (P<0.05). The findings suggest that mu-opioid receptors may be involved in the persistence of inflammatory hyperalgesia and that a delta-opioid receptor-mediated compensatory mechanism in the absence of the mu-opioid receptor is activated by persistent hyperalgesia.


Assuntos
Analgesia , Hiperalgesia/etiologia , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/farmacologia
18.
J Altern Complement Med ; 5(5): 429-36, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10537242

RESUMO

OBJECTIVE: To evaluate the effects of four herbal medicine extracts on a rat model of inflammatory hyperalgesia. DESIGN/INTERVENTIONS: Inflammation was induced by injecting complete Freund's adjuvant (CFA) into one hindpaw of each rat. Four herbs that are routinely prescribed in Traditional Chinese Medicine for treatment of pain were used: Duhuo (Radix Angelicae Pubescentis), Bai jiang cao (Patriniae Herba cum Radice), Yan hu suo (Rhizoma Corydalis) and Sanqui (Panax Notoginseng). The crude water extracts of the herbs were inected intraperitoneally following a repeated treatment profile. OUTCOME MEASURES: Thermal hyperalgesia was assessed by testing each rat's paw withdrawal response to a noxious thermal stimulus. The magnitude of edema was determined by measuring the maximal thickness of the paw with a caliper. The effect of herb extracts on motor performance was assessed by using an accelerating rotarod test. RESULT: Duhuo, Bai jiang cao, and Yan hu suo significantly attenuated CFA-induced hyperalgesia at 2 hours and facilitated the recovery from hyperalgesia (p < 0.05), when compared to saline-treated rats. The CFA-induced edema was reduced by Duhuo at 24 hours, 72 hours and 168 hours; Bai jiang cao at 24 hours, and Yan hu suo at 24 hours and 168 hours. Sanqi did not produce any significant effect on inflammation and hyperalgesia. The rotarod performance was slightly reduced by Bai jiang cao, Yan hu suo, and Sanqi (p < 0.05) but not by Duhuo treatment. CONCLUSION: The present study identified Duhuo as a selective and effective herbal agent in attenuating persistent hindpaw inflammation and hyperalgesia in rats. These results indicate that some herbal agents may provide an alternative approach to the treatment of persistant inflammatory pain and hyperalgesia.


Assuntos
Adjuvante de Freund/efeitos adversos , Hiperalgesia , Inflamação/induzido quimicamente , Extratos Vegetais , Plantas Medicinais , Animais , Ratos
19.
Brain Res ; 845(2): 165-75, 1999 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-10536195

RESUMO

The effects of vagotomy and adrenalectomy on the expression of Fos protein in brainstem neurons following the inflammation of masseter muscle were examined in order to differentiate the Fos activation related to nociceptive processing in contrast to that due to somatoautonomic processing. The inflammation was induced by a unilateral injection of complete Freund's adjuvant (CFA) into the masseter muscle under methohexital anesthesia after a small skin-cut (S-cut). After the CFA injection, Fos positive neurons were identified in bilateral spinal trigeminal nucleus (VSP), nucleus tractus solitarius (NTS), ventrolateral medulla (VLM) and inferior medial olivary nucleus (IOM). At the level of the trigeminal subnucleus interpolaris/caudalis (Vi/Vc) transition zone, there was a selective induction of Fos-like immunoreactivity (LI) in the VSP and NTS, when compared to control rats (anesthesia with or without S-cut). A major portion of the Fos-LI in the VSP at the level of the caudal Vc was apparently activated by S-cut. Bilateral adrenalectomy or a unilateral vagotomy resulted in a selective reduction of inflammation-induced Fos-LI in the VSP at the Vi/Vc transition zone (P<0.05) and NTS (P<0.05), but had less effect on Fos-LI in the caudal Vc. These results suggest that the inflammation of the masseter muscle, an injury of orofacial deep tissue, results in a widespread change in neuronal activity in the VSP and NTS that depends in part on the integrity of the adrenal cortex and vagus. Thus, in addition to somatotopically organized nociceptive responses, orofacial deep tissue injury also is coupled to somatovisceral and somatoautonomic processing that contribute to central neural activation.


Assuntos
Glândulas Suprarrenais/fisiologia , Dor Facial/metabolismo , Músculo Masseter/inervação , Proteínas Proto-Oncogênicas c-fos/biossíntese , Núcleos do Trigêmeo/metabolismo , Nervo Vago/fisiologia , Glândulas Suprarrenais/cirurgia , Adrenalectomia , Animais , Dor Facial/induzido quimicamente , Adjuvante de Freund , Masculino , Miosite/induzido quimicamente , Miosite/metabolismo , Neurônios/química , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/fisiologia , Núcleos do Trigêmeo/citologia , Vagotomia , Nervo Vago/cirurgia , Redução de Peso
20.
J Neurophysiol ; 82(3): 1244-53, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10482744

RESUMO

Studies at spinal levels indicate that peripheral tissue or nerve injury induces a state of hyperexcitability of spinal dorsal horn neurons that participates in the development of persistent pain and hyperalgesia. It has not been demonstrated that persistent injury in the orofacial region leads to a similar state of central hyperexcitability in the trigeminal system. The purpose of the present study was to conduct a parametric analysis of the response properties of nociceptive and nonnociceptive neurons in trigeminal nucleus caudalis (medullary dorsal horn, MDH) in a rat model of persistent orofacial inflammation. Neurons were recorded extracellularly and classified as low-threshold mechanoreceptive (LTM, n = 49), wide dynamic range (WDR, n = 82), and nociceptive-specific (NS, n = 11) neurons according to their response properties to mechanical stimuli applied to their cutaneous receptive fields (RFs). The inflammation was induced 24 h before the recordings by injecting complete Freund's adjuvant (CFA) into the temporomandibular joint (TMJ) capsule or the perioral (PO) skin. The mean areas of the high-threshold RFs of WDR neurons in TMJ (8.66 +/- 0.61 cm(2), n = 25) and PO (5.61 +/- 2.07 cm(2), n = 25) inflamed rats were significantly larger than those in naive rats (1.10 +/- 0. 16 cm(2), n = 32). The mean RF size in TMJ-inflamed rats also was significantly larger than that in PO-inflamed rats (P < 0.01). Furthermore the mean area of the RFs of NS neurons (3.74 +/- 1.44 cm(2), n = 5) was significantly larger in TMJ inflamed rats as compared with naive rats (0.4 +/- 0.09 cm(2), n = 3) (P < 0.05). The background activity in the TMJ- and PO-inflamed rats was generally greater in WDR and NS neurons, but less in LTM neurons, when compared with naive rats. The responses of WDR neurons to noxious mechanical stimuli were increased significantly in TMJ-inflamed rats (P < 0.05) as compared with naive rats. WDR neuronal responses to mechanical stimulation also were increased in PO-inflamed rats but to a lesser extent than in TMJ-inflamed rats. The injection of CFA into the TMJ or PO skin resulted in reduced responses of LTM neurons to mechanical stimuli. The responses of MDH nociceptive neurons to 48-55 degrees C heating were greater in inflamed rats as compared with naive rats. A subpopulation of WDR neurons recorded from TMJ (n = 4 of 10)- or PO (n = 3 of 13)-injected rats responded to cooling in addition to heating of the RFs but did not grade their responses with changes in stimulus intensity. These results indicate that persistent orofacial inflammation produced hyperexcitability of MDH nociceptive neurons. TMJ inflammation resulted in more robust changes in MDH nociceptive neurons as compared with PO inflammation, consistent with previous studies of increased inflammation, increased MDH Fos-protein expression, and increased MDH preprodynorphin mRNA expression in this deep tissue orofacial model of pain and hyperalgesia. The inflammation-induced MDH hyperexcitability may contribute to mechanisms of persistent pain associated with orofacial deep tissue painful conditions.


Assuntos
Artrite/fisiopatologia , Bulbo/fisiopatologia , Neurônios/fisiologia , Articulação Temporomandibular , Animais , Artrite/patologia , Dermatite/patologia , Dermatite/fisiopatologia , Eletrofisiologia , Face , Temperatura Alta , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Bulbo/patologia , Doenças da Boca/patologia , Doenças da Boca/fisiopatologia , Nociceptores/fisiologia , Estimulação Física , Ratos , Ratos Sprague-Dawley , Tempo de Reação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA