Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
J Dent Res ; 97(5): 483-491, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29328868

RESUMEN

For decades, dental schools in the United States have endured a significant faculty shortage. Studies have determined that the top 2 sources of dental faculty are advanced education programs and private practice. Those who have completed both DDS and PhD training are considered prime candidates for dental faculty positions. However, there is no national database to track those trainees and no evidence to indicate that they entered academia upon graduation. The objective of this study was to assess outcomes of dental school-affiliated oral sciences PhD program enrollment, graduates, and placement between 1994 and 2016. Using the American Dental Association annual survey of advanced dental education programs not accredited by the Commission on Dental Accreditation and data obtained from 22 oral sciences PhD programs, we assessed student demographics, enrollment, graduation, and placement. Based on the data provided by program directors, the average new enrollment was 33, and graduation was 26 per year. A total of 605 graduated; 39 did not complete; and 168 were still in training. Among those 605 graduates, 211 were faculty in U.S. academic institutions, and 77 were faculty in foreign institutions. Given that vacant budgeted full-time faculty positions averaged 257 per year during this period, graduates from those oral sciences PhD programs who entered academia in the United States would have filled 9 (3.6%) vacant faculty positions per year. Therefore, PhD programs have consistently generated only a small pipeline of dental school faculty. Better mentoring to retain talent in academia is necessary. Stronger support and creative funding plans are essential to sustain the PhD program. Furthermore, the oral sciences PhD program database should be established and maintained by dental professional organizations to allow assessments of training models, trends of enrollment, graduation, and placement outcomes.


Asunto(s)
Educación de Posgrado en Odontología/estadística & datos numéricos , Humanos , Facultades de Odontología/estadística & datos numéricos , Encuestas y Cuestionarios , Estados Unidos
2.
J Dent Res ; 95(10): 1117-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27307050

RESUMEN

Acute or chronic inflammation comprises a highly prevalent type of orofacial pain and is mediated by the generation of endogenous agonists that activate numerous receptors expressed on terminals of trigeminal (TG) nociceptive afferent neurons. One such studied receptor is transient receptor potential vanilloid subtype 1 (TRPV1). TRPV1 is a ligand-gated cation channel that is expressed on a major subclass of nociceptors and is found in many orofacial tissues, including dental pulp. Antagonists to TRPV1 reveal an important role for this channel in mediating hypersensitivity in preclinical models of inflammatory or neuropathic pain. Recent studies have demonstrated that endogenous TRPV1 agonists are generated by oxidation of omega-6 polyunsaturated fatty acids, including both linoleic acid and arachidonic acid. A major mechanism triggering the release of oxidative linoleic acid metabolites (OLAMs) and oxidative arachidonic acid metabolites (OAAMs) is the action of oxidative enzymes. Oxidative enzymes such as cytochrome P450 isozymes are rapidly upregulated in TG neurons after orofacial inflammation and increase the capacity of TG neurons to generate OLAMs. Cytochrome P450 isozymes are also increased in immune cells in irreversibly inflamed human dental pulp, and extracts of this tissue have significantly increased capacity to generate OLAMs. Together, these studies point to a novel pain mechanism involving the enzymatic generation of endogenous OLAM and OAAM agonists of TRPV1. This finding provides a rationale for an entirely new class of analgesics by inhibition of oxidative enzyme activity.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Dolor Facial/metabolismo , Ácidos Grasos Omega-6/metabolismo , Nociceptores/metabolismo , Canales Catiónicos TRPV/agonistas , Inhibidores Enzimáticos/farmacología , Humanos , Hiperalgesia/metabolismo , Inflamación/metabolismo , Isoenzimas/metabolismo , Oxidación-Reducción
3.
J Dent Res ; 93(12): 1296-303, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25201919

RESUMEN

Creating an optimal microenvironment that mimics the extracellular matrix (ECM) of natural pulp and securing an adequate blood supply for the survival of cell transplants are major hurdles that need to be overcome in dental pulp regeneration. However, many currently available scaffolds fail to mimic essential functions of natural ECM. The present study investigated a novel approach involving the use of scaffold-free microtissue spheroids of dental pulp stem cells (DPSCs) prevascularized by human umbilical vein endothelial cells (HUVECs) in pulp regeneration. In vitro-fabricated microtissue spheroids were inserted into the canal space of tooth-root slices and were implanted subcutaneously into immunodeficient mice. Histological examination revealed that, after four-week implantation, tooth-root slices containing microtissue spheroids resulted in well-vascularized and cellular pulp-like tissues, compared with empty tooth-root slices, which were filled with only subcutaneous fat tissue. Immunohistochemical staining indicated that the tissue found in the tooth-root slices was of human origin, as characterized by the expression of human mitochondria, and contained odontoblast-like cells organized along the dentin, as assessed by immunostaining for nestin and dentin sialoprotein (DSP). Vascular structures formed by HUVECs in vitro were successfully anastomosed with the host vasculature upon transplantation in vivo, as shown by immunostaining for human CD31. Collectively, these findings demonstrate that prevascularized, scaffold-free, microtissue spheroids can successfully regenerate vascular dental pulp-like tissue and also highlight the significance of the microtissue microenvironment as an optimal environment for successful pulp-regeneration strategies.


Asunto(s)
Pulpa Dental/citología , Regeneración/fisiología , Esferoides Celulares/fisiología , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Adolescente , Adulto , Animales , Técnicas de Cultivo de Célula , Microambiente Celular/fisiología , Técnicas de Cocultivo , Pulpa Dental/fisiología , Cavidad Pulpar/citología , Dentina/citología , Proteínas de la Matriz Extracelular , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Ratones SCID , Microvasos/citología , Mitocondrias/ultraestructura , Modelos Animales , Neovascularización Fisiológica/fisiología , Nestina/análisis , Odontoblastos/citología , Fosfoproteínas , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Sialoglicoproteínas , Trasplante de Células Madre/métodos , Adulto Joven
4.
Neuroscience ; 203: 207-15, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22209919

RESUMEN

The peripheral serotonergic system has been implicated in the modulation of an array of pain states, from migraine to fibromyalgia; however, the mechanism by which serotonin (5HT) induces pain is unclear. Peripherally released 5HT induces thermal hyperalgesia, possibly via modulation of the transient receptor potential V1 (TRPV1) channel, which is gated by various noxious stimuli, including capsaicin. We previously reported in vitro that 5HT increases calcium accumulation in the capsaicin-sensitive population of sensory neurons with a corresponding increase in proinflammatory neuropeptide release, and both are antagonized by pretreatment with 5HT(2A) and 5HT(3) antagonists, as well as the anti-migraine drug sumatriptan. In the current study, we extended these findings in vivo using the rat hind paw thermal assay to test the hypothesis that peripheral 5HT enhances TRPV1-evoked thermal hyperalgesia that can be attenuated with 5HT(2A) and 5HT(3) receptor antagonists, as well as sumatriptan. Thermal hyperalgesia and edema were established by 5HT injection (0.1-10 nmol/100 µl) into the rat hind paw, and the latency to paw withdrawal (PWL) from noxious heat was determined. Rats were then pretreated with either 5HT before capsaicin (3 nmol/10 µl), the 5HT(2A) receptor antagonist ketanserin or the 5HT(3) receptor antagonist granisetron (0.0001-0.1 nmol/100 µl) before 5HT and/or capsaicin, or the 5HT(1B/1D) receptor agonist sumatriptan (0.01-1 nmol/100 µl) before capsaicin, and PWL was determined. We report that 5HT pretreatment enhances TRPV1-evoked thermal hyperalgesia, which is attenuated with local pretreatment with ketanserin, granisetron, or sumatriptan. We also report that peripheral 5HT induced a similar magnitude of thermal hyperalgesia in male and female rats. Overall, our results provide in vivo evidence supporting an enhancing role of 5HT on TRPV1-evoked thermal hyperalgesia, which can be attenuated by peripheral serotonergic intervention.


Asunto(s)
Capsaicina/farmacología , Hiperalgesia/tratamiento farmacológico , Ketanserina/uso terapéutico , Serotonina/farmacología , Sumatriptán/uso terapéutico , Animales , Femenino , Hiperalgesia/inducido químicamente , Masculino , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología
5.
J Dent Res ; 90(6): 759-64, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21393555

RESUMEN

Recent studies have demonstrated that the lipopolysaccharide (LPS) receptor (TLR4) is expressed in TRPV1 containing trigeminal sensory neurons. In this study, we evaluated whether LPS activates trigeminal neurons, and sensitizes TRPV1 responses via TLR4. To test this novel hypothesis, we first demonstrated that LPS binds to receptors in trigeminal neurons using competitive binding. Second, we demonstrated that LPS evoked a concentration-dependent increase in intracellular calcium accumulation (Ca(2+))(i) and inward currents. Third, LPS significantly sensitized TRPV1 to capsaicin measured by (Ca(2+))(i), release of calcitonin gene-related peptide, and inward currents. Importantly, a selective TLR4 antagonist blocked these effects. Analysis of these data, collectively, demonstrates that LPS is capable of directly activating trigeminal neurons, and sensitizing TRPV1 via a TLR4-mediated mechanism. These findings are consistent with the hypothesis that trigeminal neurons are capable of detecting pathogenic bacterial components leading to sensitization of TRPV1, possibly contributing to the inflammatory pain often observed in bacterial infections.


Asunto(s)
Lipopolisacáridos/metabolismo , Nociceptores/metabolismo , Canales Catiónicos TRPV/metabolismo , Receptor Toll-Like 4/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Unión Competitiva , Péptido Relacionado con Gen de Calcitonina/biosíntesis , Señalización del Calcio , Capsaicina/farmacología , Células Cultivadas , Masculino , Nociceptores/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Fármacos del Sistema Sensorial/farmacología , Receptor Toll-Like 4/agonistas , Ganglio del Trigémino/citología
6.
J Dent Res ; 87(10): 948-52, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18809749

RESUMEN

Neuropeptide Y (NPY) is an important modulatory neuropeptide that regulates several physiological systems, including the activity of sensory neurons. We evaluated whether activation of the NPY Y1 receptor could modulate the activity of capsaicin-sensitive nociceptors in trigeminal ganglia and dental pulp. We tested this hypothesis by measuring capsaicin-stimulated calcitonin gene-related peptide release (CGRP) as a measure of nociceptor activity. Capsaicin-evoked CGRP release was inhibited by 50% (p < 0.05) in trigeminal ganglia and by 26% (p < 0.05) in dental pulp when tissues were pre-treated with [Leu(31),Pro(34)]NPY. The Y1 receptor was found to co-localize with the capsaicin receptor TRPV1 in trigeminal ganglia. These results demonstrate that activation of the Y1 receptor results in the inhibition of the activity of capsaicin-sensitive nociceptors in the trigeminal ganglia and dental pulp. These findings are relevant to the physiological modulation of dental nociceptors by endogenous NPY and demonstrate an important novel analgesic target for the treatment of dental pain.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/biosíntesis , Pulpa Dental/inervación , Nociceptores/efectos de los fármacos , Receptores de Neuropéptido Y/fisiología , Odontalgia/tratamiento farmacológico , Animales , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Capsaicina/farmacología , Pulpa Dental/efectos de los fármacos , Pulpa Dental/metabolismo , Masculino , Inflamación Neurogénica/tratamiento farmacológico , Nociceptores/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/administración & dosificación , Canales Catiónicos TRPV/antagonistas & inhibidores , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo
7.
Neuroscience ; 155(2): 503-9, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18582539

RESUMEN

Tumor necrosis factor alpha (TNFalpha), a pro-inflammatory cytokine, enhances the development of pain and hyperalgesia, although the molecular mechanisms are not well understood. This study evaluated the hypothesis that TNFalpha increases the sensitivity of rat trigeminal neurons to capsaicin via two different mechanisms triggered by either brief or sustained exposure to the cytokine. A brief (5 min) application of TNFalpha significantly sensitized capsaicin-evoked accumulation of intracellular calcium ([Ca2+]i) (226.4+/-37.7 nM vs. 167.5+/-31.3 nM) and increased capsaicin-evoked nocifensive behavior (78.3+/-9.7 vs. 30.9+/-3.6 s) as compared with vehicle pretreatment (P<0.01 for both). Sustained (30 min to 4 h) exposure of cultured neurons to TNFalpha evoked a twofold increase in mRNA transcript (P<0.05) and protein levels (P<0.01) of transient potential receptor vanilloid type 1 (TRPV1). This long-term up-regulation of TRPV1 expression by TNFalpha correlated with enhancement in capsaicin-induced calcitonin gene-related peptide release (P<0.05). Demonstration of colocalization of TNFalpha receptor subtypes I and II with TRPV1 in almost all (>90%) TRPV1 expressing neurons provides evidence consistent with a direct interaction on the same subpopulation of sensory neurons. In summary, our data demonstrate that TNFalpha directly enhances the sensitivity of rat trigeminal neurons to capsaicin via both rapid, non-genomic mechanisms as well as sustained genomic regulation in TRPV1 expression. Thus, increased sensitization and up-regulation of TRPV1 constitutes a potential mechanism by which TNFalpha mediates inflammatory hyperalgesia and pain.


Asunto(s)
Capsaicina/farmacología , Neuronas/efectos de los fármacos , Nervio Trigémino/citología , Neuralgia del Trigémino/fisiopatología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Calcio/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Expresión Génica/efectos de los fármacos , Masculino , Neuronas/citología , Neuronas/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Fármacos del Sistema Sensorial/farmacología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Tiempo , Regulación hacia Arriba/efectos de los fármacos
8.
J Dent Res ; 87(3): 262-6, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18296611

RESUMEN

Orofacial inflammation is associated with prostaglandin release and the sensitization of nociceptive receptors such as the transient receptor potential subtype V(1) (TRPV(1)). We hypothesized that certain PGE(2) receptor subtypes (EP1-EP4) are co-expressed with TRPV(1) in trigeminal nociceptors and sensitize responses to a TRPV(1) agonist, capsaicin. Accordingly, combined in situ hybridization was performed with immunohistochemistry on rat trigeminal ganglia. We next evaluated the effects of specific EP2 and EP3 agonists (butaprost and sulprostone) in cultured trigeminal ganglia neurons. The results showed that EP2 and EP3 are expressed in trigeminal neurons (58% and 53% of total neurons, respectively) and are co-expressed in TRPV(1)-positive neurons (64% and 67 % of TRPV(1)-positive neurons, respectively). Moreover, most of the cells expressing EP2 or EP3 mRNA were of small to medium diameter (< 30 microm). The application of butaprost and sulprostone triggered neuropeptide exocytosis, and butaprost sensitized capsaicin responses. Analysis of these data, collectively, supports the hypothesis that prostaglandins regulate trigeminal TRPV(1) nociceptors via activation of the EP2 and EP3 receptors.


Asunto(s)
Nociceptores/metabolismo , Receptores de Prostaglandina E/metabolismo , Ganglio del Trigémino/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Capsaicina/farmacología , Tamaño de la Célula , Células Cultivadas , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Exocitosis/efectos de los fármacos , Inmunohistoquímica , Hibridación in Situ , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/metabolismo , Nociceptores/efectos de los fármacos , Prostaglandinas E Sintéticas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP3 de Receptores de Prostaglandina E , Canales Catiónicos TRPV/agonistas , Ganglio del Trigémino/efectos de los fármacos
9.
J Dent Res ; 86(6): 550-5, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17525356

RESUMEN

The transient receptor potential ankyrin repeat 1 (TRPA1) channel is believed to be involved in many forms of acute and chronic hyperalgesia. Nerve Growth Factor (NGF) regulates chronic inflammatory hyperalgesia by controlling gene expression in sensory neurons, including genes involved in inflammatory hyperalgesia in the dental pulp. We hypothesized that NGF increases functional activities of the TRPA1 channel in trigeminal ganglion neurons. Here, we show that NGF induced a concentration- and time-dependent up-regulation of TRPA1 mRNA in trigeminal ganglia neurons, as detected by real-time RT-PCR and in situ hybridization. In addition, NGF evoked a time-dependent increase of mustard oil (MO)-evoked TRPA1 activation in trigeminal ganglia neurons. Collectively, these findings demonstrate that NGF participates in the functional up-regulation of TRPA1 in trigeminal ganglia neurons. These enhanced activities of TRPA1 could play an important role in the development of hyperalgesia following nerve injury and inflammation in the orofacial region.


Asunto(s)
Repetición de Anquirina/efectos de los fármacos , Canales de Calcio/efectos de los fármacos , Dolor Facial/fisiopatología , Factor de Crecimiento Nervioso/farmacología , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Ancirinas , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/fisiopatología , Inmunohistoquímica , Hibridación in Situ , Masculino , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canal Catiónico TRPA1 , Canales Catiónicos TRPC , Factores de Tiempo , Ganglio del Trigémino/efectos de los fármacos
10.
Neuroscience ; 144(3): 889-97, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17157995

RESUMEN

The binding of integrins to the extracellular matrix results in focal organization of the cytoskeleton and the genesis of intracellular signals that regulate vital neuronal functions. Recent evidence suggests that integrins modulate G-protein-coupled receptor (GPCR) signaling in hippocampal neurons. In this study we evaluated the hypothesis that integrins regulate the mu opioid receptor in rat trigeminal ganglion neurons. For these studies, primary cultures of adult rat trigeminal ganglion neurons were used to demonstrate the colocalization of beta1 and beta3 integrins with mu opioid receptor in caveolin-1-rich membrane fractions, and at focal adhesions sites generated by integrin ligand binding. Furthermore, we show that the mu opioid receptor agonist, DAMGO ([D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin), inhibits cyclic AMP (cAMP) accumulation in response to prostaglandin E2 (PGE(2)) stimulation in bradykinin-primed, but not unprimed, cultured trigeminal ganglia neurons. Application of soluble GRGDS (Gly-Arg-Gly-Asp-Ser) peptides that bind specific integrins (i.e. RGD-binding integrins) completely abolished the DAMGO effect in bradykinin-primed trigeminal ganglia neurons, but did not alter bradykinin-mediated hydrolysis of phosphatidylinositol. Likewise, monospecific anti-beta1 and anti-beta3 integrin subunit antibodies blocked this DAMGO effect in bradykinin-primed trigeminal ganglia neurons. Indeed, application of anti-beta1 integrin subunit actually reversed DAMGO signaling, resulting in increased cAMP accumulation in these cells. This suggests that the relative amounts of specific activated integrins at focal adhesions may govern signaling by the mu opioid receptor, perhaps by altering interactions with G proteins (e.g. Galphai vs. Galphas). Collectively, these data provide the first evidence that specific integrins regulate opioid receptor signaling in sensory neurons.


Asunto(s)
Integrinas/metabolismo , Neuronas Aferentes/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal/fisiología , Ganglio del Trigémino/metabolismo , Analgésicos Opioides/farmacología , Animales , Anticuerpos/farmacología , Bradiquinina/metabolismo , Bradiquinina/farmacología , Células Cultivadas , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Adhesiones Focales/metabolismo , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Masculino , Neuronas Aferentes/efectos de los fármacos , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Ganglio del Trigémino/citología , Ganglio del Trigémino/efectos de los fármacos
11.
Br J Pharmacol ; 150(1): 72-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143304

RESUMEN

BACKGROUND AND PURPOSE: Although previous studies have demonstrated that neuropeptide Y (NPY) modulates nociceptors, the relative contributions of the Y1 and Y2 receptors are unknown. Therefore, we evaluated the effect of Y1 and Y2 receptor activation on nociceptors stimulated by bradykinin (BK) and prostaglandin E2 (PGE2). EXPERIMENTAL APPROACH: Combined immunohistochemistry (IHC) with in situ hybridization (ISH) demonstrated that Y1- and Y2-receptors are collocated with bradykinin (2) (B2)-receptors in rat trigeminal ganglia (TG). The relative functions of the Y1 and Y2 receptors in modulating BK/PGE2-evoked CGRP release and increased intracellular calcium levels in cultured TG neurons were evaluated. KEY RESULTS: The Y1 and Y2 receptors are co-expressed with B2 in TG neurons, suggesting the potential for direct NPY modulation of BK responses. Pretreatment with the Y1 agonist [Leu31,Pro34]-NPY, inhibited BK/PGE2-evoked CGRP release. Conversely, pretreatment with PYY(3-36), a Y2 agonist, increased BK/PGE2 evoked CGRP release. Treatment with NPY evoked an overall inhibitory effect, although of lesser magnitude. Similarly, [Leu31,Pro34]-NPY inhibited BK/PGE2-evoked increases in intracellular calcium levels whereas PYY(3-36) increased responses. NPY inhibition of BK/PGE2-evoked release of CGRP was reversed by the Y1 receptor antagonist, BIBO3304, and higher concentrations of BIBO3304 significantly facilitated CGRP release. The Y2 receptor antagonist, BIIE0246, enhanced the inhibitory NPY effects. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that NPY modulation of peptidergic neurons is due to net activation of inhibitory Y1 and excitatory Y2 receptor systems. The relative expression or activity of these opposing receptor systems may mediate dynamic responses to injury and pain.


Asunto(s)
Bradiquinina/farmacología , Dinoprostona/farmacología , Neuropéptido Y/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Neuropéptido Y/efectos de los fármacos , Receptores de Neuropéptido/efectos de los fármacos , Nervio Trigémino/efectos de los fármacos , Animales , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratas , Ratas Sprague-Dawley
12.
J Dent Res ; 85(11): 1046-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17062748

RESUMEN

The TRPV1 receptor acts as a sensor for environmental changes in pH and temperature. Since many nociceptors express TRPV1, it is possible that local tissue-cooling may inhibit nociceptor activity via reduction of TRPV1 activation. The present study used isolated superfused rat dental pulp to test the hypothesis that capsaicin receptors are activated in inflamed tissue, as measured by alterations in neuropeptide release. We tested the hypothesis that alterations in the tissue temperature and pH of isolated superfused rat dental pulp regulate capsaicin-induced release of calcitonin gene-related peptide (CGRP). Application of capsaicin with increased proton concentration (i.e., lowered pH) produced a nearly two-fold increase in peak immunoreactive CGRP release, as compared with capsaicin applied at a pH of 7.4. Reduction in tissue temperature from 37 degrees C to 26 degrees C completely blocked the capsaicin effect. The study indicates that environmental stimuli regulate the activity of capsaicin-sensitive neurons innervating dental pulp, and these factors may be significant clinically in the development and amelioration of dental pain.


Asunto(s)
Temperatura Corporal/fisiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Pulpa Dental/inervación , Nociceptores/fisiología , Odontalgia/fisiopatología , Analgésicos no Narcóticos/farmacología , Animales , Capsaicina/farmacología , Concentración de Iones de Hidrógeno , Nociceptores/efectos de los fármacos , Pulpitis/fisiopatología , Ratas , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo
13.
Neuroscience ; 141(4): 2107-16, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16809002

RESUMEN

Neuronal proteins have been traditionally viewed as being derived solely from the soma; however, accumulating evidence indicates that dendritic and axonal sites are capable of a more autonomous role in terms of new protein synthesis. Such extra-somal translation allows for more rapid, on-demand regulation of neuronal structure and function than would otherwise be possible. While mechanisms of dendritic RNA transport have been elucidated, it remains unclear how RNA is trafficked into the axon for this purpose. Primary afferent neurons of the dorsal root (DRG) and trigeminal (TG) ganglia have among the longest axons in the neuraxis and such axonal protein synthesis would be advantageous, given the greater time involved for protein trafficking to occur via axonal transport. Therefore, we hypothesized that these primary sensory neurons might express proteins involved in RNA transport. Rat DRG and TG neurons expressed staufen (stau) 1 and 2 (detected at the mRNA level) and stau2 and fragile x mental retardation protein (FMRP; detected at the protein level). Stau2 mRNA was also detected in human TG neurons. Stau2 and FMRP protein were localized to the sciatic nerve and dorsal roots by immunohistochemistry and to dorsal roots by Western blot. Stau2 and FMRP immunoreactivities colocalized with transient receptor potential channel type 1 immunoreactivity in sensory axons of the sciatic nerve and dorsal root, suggesting that these proteins are being transported into the peripheral and central terminals of nociceptive sensory axons. Based on these findings, we propose that stau2 and FMRP proteins are attractive candidates to subserve RNA transport in sensory neurons, linking somal transcriptional events to axonal translation.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Ganglios Espinales/citología , Neuronas Aferentes/metabolismo , Proteínas de Unión al ARN/metabolismo , Ganglio del Trigémino/citología , Animales , Northern Blotting/métodos , Western Blotting/métodos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Expresión Génica/fisiología , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Técnicas In Vitro , Masculino , Proteínas de Unión al ARN/clasificación , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley
14.
J Dent Res ; 85(1): 49-53, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16373680

RESUMEN

Although certain bacterial species appear to be risk factors for pain due to odontogenic infections, comparatively little is known about the potential mechanisms mediating this effect. In this study, we tested the hypothesis that trigeminal nociceptive neurons express the TLR4 or CD14 receptors, thus enabling sensory neurons to detect and respond to tissue levels of bacterial substances such as lipopolysaccharide (LPS). Immunohistochemical analyses of human and rat trigeminal neurons demonstrated that a capsaicin-sensitive subclass of nociceptors (defined by expression of TRPV1, a capsaicin receptor) expresses both TLR4 and CD14. Moreover, human dental pulp collected from patients with caries lesions demonstrated co-localization of TLR4 and CD14, with markers of peripheral sensory neurons. Collectively, these studies indicate that the capsaicin-sensitive subclass of trigeminal nociceptors expresses TLR4 and CD14. These results indicate that pain due to bacterial infections may result, in part, from direct activation of nociceptors by bacterial products such as LPS.


Asunto(s)
Infecciones Bacterianas/patología , Receptores de Lipopolisacáridos/análisis , Nociceptores/citología , Dolor/patología , Receptor Toll-Like 4/análisis , Ganglio del Trigémino/citología , Anciano , Animales , Cadáver , Pulpa Dental/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Modelos Animales , Neuronas Aferentes/citología , Pulpitis/patología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/análisis
15.
Neuroscience ; 125(3): 703-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15099684

RESUMEN

Neuropeptide Y (NPY) is expressed in certain primary afferent fibers, is up-regulated in response to tissue injury and is capable of inhibiting nociceptive behavior at the spinal level. However, the spinal mechanism(s) for NPY-evoked antinociception is unknown. In this study, we evaluated the hypothesis that agonists at the NPY Y1 receptor subtype (Y1-R) inhibit exocytosis from the capsaicin-sensitive class of nociceptors. Using in vitro superfusion of rat dorsal spinal cord slices, pre-treatment with the Y1-R agonist [Leu(31)Pro(34)]NPY significantly inhibited capsaicin-evoked release of immunoreactive calcitonin gene-related peptide with an EC(50) value of 10.6 nM. This inhibitory effect was concentration dependent, significantly attenuated by pre-treatment with the Y1 receptor antagonist BIBP3226 and reproduced by synthetic NPY. Examination of adult rat dorsal root ganglia using double immunofluorescent labeling revealed frequent co-localization of Y1 receptor immunoreactivity in vanilloid receptor type 1-immunoreactive neurons, indicating that Y1 agonists may directly modulate the capsaicin-sensitive class of nociceptors. Collectively, these results indicate that NPY is capable of inhibiting capsaicin-sensitive neurons via a Y1 receptor mechanism, suggesting the mechanisms for spinal NPY-induced antinociception is due, at least in part, to inhibition of central terminals of capsaicin-sensitive nociceptors.


Asunto(s)
Analgésicos/farmacología , Arginina/análogos & derivados , Ganglios Espinales/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuropéptido Y/farmacología , Nociceptores/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Animales , Arginina/farmacología , Péptido Relacionado con Gen de Calcitonina/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/antagonistas & inhibidores , Capsaicina/farmacología , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/citología , Técnicas In Vitro , Masculino , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Neuropéptido Y/análogos & derivados , Neuropéptido Y/metabolismo , Nociceptores/metabolismo , Fragmentos de Péptidos/farmacología , Células del Asta Posterior/citología , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Droga/metabolismo , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo
16.
Peptides ; 24(8): 1163-74, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14612187

RESUMEN

Before parturition the uterine cervix undergoes a ripening process ("softens" and dilates) to allow passage of the fetus at term. The exact mechanism(s) responsible for cervical ripening are unknown, though a role for peptidergic sensory neurons is emerging. Previous work demonstrated that administration of substance P (SP) to ovariectomized rats caused events associated with cervical ripening, that production of SP in cervix-related dorsal root ganglion (DRG) is estrogen responsive, and that release of SP from neurons terminating in the cervix and spinal cord peaks prior to parturition. The present study was designed to test the hypothesis that calcitonin gene-related peptide (CGRP), a neuropeptide co-stored with SP in many sensory neurons, undergoes changes with pregnancy and hormonal environment. Immunohistochemistry, in situ hybridization, reverse transcriptase-polymerase chain reaction (RT-PCR) and radioimmunoassay (RIA) were used to investigate CGRP in L6-S1 DRG, spinal cord and cervix during pregnancy and the role of estrogen in CGRP synthesis. CGRP-immunoreactive primary sensory neurons expressed estrogen receptors (ER-alpha and ER-beta). In the cervix, CGRP concentrations decreased, but in the L6-S1 DRG and the spinal cord segments, CGRP levels increased, with peak effects observed at day 20 of gestation. CGRP mRNA synthesis increased in DRG over pregnancy. Sensory neurons of ovariectomized rats treated with estrogen showed increased CGRP mRNA synthesis in a dose-related manner, an effect blocked by the ER antagonist ICI 182 780. From these results, we postulate that synthesis of CGRP in L6-S1 DRG and utilization in the cervix increase over pregnancy and this synthesis is the under influence of the estrogen-ER system. Collectively, these data are consistent with the hypothesis that CGRP plays a role in cervical ripening and, consequently in the birth process.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/genética , Cuello del Útero/fisiología , Estrógenos/fisiología , Ganglios Espinales/fisiología , Médula Espinal/fisiología , Animales , Péptido Relacionado con Gen de Calcitonina/análisis , Péptido Relacionado con Gen de Calcitonina/biosíntesis , Femenino , Inmunohistoquímica , Hibridación in Situ , Embarazo , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley
17.
Neuroscience ; 120(1): 155-62, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12849749

RESUMEN

Cannabinoid compounds have been shown to produce antinociception and antihyperalgesia by acting upon cannabinoid receptors located in both the CNS and the periphery. A potential mechanism by which cannabinoids could inhibit nociception in the periphery is the activation of cannabinoid receptors located on one or more classes of primary nociceptive neurons. To address this hypothesis, we evaluated the neuronal distribution of cannabinoid receptor type 1 (CB1) in the trigeminal ganglion (TG) of the adult rat through combined in situ hybridization (ISH) and immunohistochemistry (IHC). CB1 receptor mRNA was localized mainly to medium and large diameter neurons of the maxillary and mandibular branches of the TG. Consistent with this distribution, in a de facto nociceptive sensory neuron population that exhibited vanilloid receptor type 1 immunoreactivity, colocalization with CB1 mRNA was also sparse (<5%). Furthermore, very few neurons (approximately 5%) in the peptidergic (defined as calcitonin gene-related peptide- or substance P-immunoreactive) or the isolectin B4-binding sensory neuron populations contained CB1 mRNA. In contrast, and consistent with the neuron-size distribution for CB1, nearly 75% of CB1-positive neurons exhibited N52-immunoreactivity, a marker of myelinated axons. These results indicate that in the rat TG, CB1 receptors are expressed predominantly in neurons that are not thought to subserve nociceptive neurotransmission in the noninjured animal. Taken together with the absence of an above background in situ signal for CB2 mRNA in TG neurons, these findings suggest that the peripherally mediated antinociceptive effects of cannabinoids may involve either as yet unidentified receptors or interaction with afferent neuron populations that normally subserve non-nociceptive functions.


Asunto(s)
Cannabinoides/análisis , Neuronas/química , Receptor Cannabinoide CB2 , Receptores de Droga/análisis , Ganglio del Trigémino/química , Animales , Masculino , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Cannabinoides
18.
J Dent Res ; 82(5): 398-401, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12709509

RESUMEN

Neurotransmission from sympathetic and peptidergic afferent fibers participates in the regulation of pulpal blood flow (PBF) via opposing effects. In this study, we directly tested the hypothesis that activation of pulpal sympathetic terminals inhibits exocytosis of immunoreactive calcitonin gene-related peptide (iCGRP) from peptidergic afferents innervating bovine dental pulp. The results demonstrate that norepinephrine inhibits capsaicin-evoked iCGRP release. The application of alpha-adrenergic antagonists (phentolamine or phenoxybenzamine) increased spontaneous release of iCGRP. Moreover, administration of agents that evoke the release of sympathetic neurotransmitters (guanethidine or reserpine) inhibited capsaicin-evoked iCGRP release. Collectively, these results indicate that sympathetic neurotransmission inhibits exocytosis from pulpal peptidergic afferent fibers. Analysis of these data supports the hypothesis that peripheral sympathetic vasomotor control may operate by a direct mechanism (vasoconstriction) as well as by an indirect mechanism (e.g., inhibition of exocytosis from afferent fibers). Since capsaicin-sensitive neurons are nociceptors, it is possible that certain sympathetic neurotransmission may modulate pain.


Asunto(s)
Fibras Adrenérgicas/fisiología , Péptido Relacionado con Gen de Calcitonina/fisiología , Pulpa Dental/irrigación sanguínea , Pulpa Dental/inervación , Vasoconstricción/fisiología , Sistema Vasomotor/fisiología , Fibras Adrenérgicas/efectos de los fármacos , Antagonistas Adrenérgicos alfa/farmacología , Análisis de Varianza , Animales , Capsaicina/farmacología , Bovinos , Exocitosis/efectos de los fármacos , Femenino , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Estadísticas no Paramétricas , Simpaticolíticos/farmacología , Nervio Trigémino/efectos de los fármacos , Vasoconstrictores/farmacología
19.
J Dent Res ; 82(4): 308-11, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12651937

RESUMEN

Previous studies have suggested that neurotransmitter substances from the sympatho-adrenomedullary system regulate pulpal blood flow (PBF), in part, by the inhibition of vasoactive neuropeptide release from pulpal sensory neurons. However, no study has evaluated the role of beta-adrenoceptors. We evaluated the hypothesis that activation of beta-adrenoceptors inhibits immunoreactive calcitonin gene-related peptide (iCGRP) release from capsaicin-sensitive nociceptive neurons via in vitro superfusion of bovine dental pulp. Either norepinephrine or epinephrine inhibited capsaicin-evoked iCGRP. The norepinephrine effect was blocked by the selective beta(2)-adrenoceptor antagonist, ICI 118,551, but not by pre-treatment with the selective beta(1)-adrenoceptor antagonist, atenolol. In addition, application of albuterol, a selective beta(2)-adrenoceptor agonist, significantly blocked capsaicin-evoked release of iCGRP. Collectively, these studies demonstrate that activation of beta(2)-adrenoceptors in dental pulp significantly reduces exocytosis of neuropeptides from capsaicin-sensitive nociceptors. This effect may have physiologic significance in regulating PBF. Moreover, since capsaicin selectively activates nociceptors, beta(2)-adrenoceptor agonists may have clinical utility as peripherally acting therapeutics for dental pain and inflammation.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2 , Péptido Relacionado con Gen de Calcitonina/metabolismo , Pulpa Dental/inervación , Receptores Adrenérgicos beta 2/fisiología , Adrenérgicos/farmacología , Albuterol/farmacología , Animales , Atenolol/farmacología , Capsaicina/farmacología , Bovinos , Epinefrina/farmacología , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Femenino , Irritantes/farmacología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Norepinefrina/farmacología , Propanolaminas/farmacología , Receptores Adrenérgicos beta 2/efectos de los fármacos
20.
Eur J Neurosci ; 14(7): 1113-20, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11683903

RESUMEN

Many of the physiological hallmarks associated with neurogenic inflammatory processes in cutaneous tissues are similarly present within orofacial structures. Such attributes include the dependence upon capsaicin-sensitive sensory neurons and the involvement of certain inflammatory mediators derived therein, including calcitonin gene-related peptide (CGRP). However, there are also important differences between the trigeminal and spinal nervous systems, and the potential contributions of neurogenic processes to inflammatory disease within the trigeminal system have yet to be fully elucidated. We present here a model system that affords the ability to study mechanisms regulating the efferent functions of peptidergic terminals that may subserve neurogenic inflammation within the oral cavity. Freshly dissected buccal mucosa tissue from adult, male, Sprague-Dawley rats was placed into chambers and superfused with oxygenated, Krebs buffer. Serial aliquots of the egressing superfusate were acquired and analysed by radioimmunoassay for immunoreactive CGRP (iCGRP). Addition of the selective excitotoxin, capsaicin (10-300 microm), to the superfusion buffer resulted in a significant, concentration-dependent increase in superfusate levels of iCGRP. Similarly, release of iCGRP from the buccal mucosa could also be evoked by a depolarizing concentration of potassium chloride (50 mm) or by the calcium ionophore A23187 (1 microm). The specific, capsaicin receptor antagonist, capsazepine (300 microm), completely abolished the capsaicin-evoked release of iCGRP while having no effect whatsoever on the potassium-evoked release. Moreover, capsaicin-evoked release was dependent upon the presence of extracellular calcium ions and was significantly, though incompletely, attenuated by neonatal capsaicin denervation. Collectively, these data indicate that the evoked neurosecretion of iCGRP in response to capsaicin occurs via a vanilloid receptor-mediated, exocytotic mechanism. The model system described here should greatly facilitate future investigations designed to identify and characterize the stimuli that regulate the release of CGRP or other neurosecretory substances in isolated tissues. This system may also be used to elucidate the role of these mediators in the aetiology of inflammatory processes within the trigeminal field of innervation.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacología , Mediadores de Inflamación/metabolismo , Mucosa Bucal/inervación , Mucosa Bucal/metabolismo , Inflamación Neurogénica/metabolismo , Nervio Trigémino/metabolismo , Animales , Bradiquinina/farmacología , Calcimicina/farmacología , Calcio/metabolismo , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Histamina/farmacología , Ionóforos/farmacología , Masculino , Mucosa Bucal/efectos de los fármacos , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/fisiopatología , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Técnicas de Cultivo de Órganos , Dimensión del Dolor/efectos de los fármacos , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Serotonina/farmacología , Nervio Trigémino/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...