Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pain ; 19: 17448069231222406, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38073552

RESUMO

Changes in sensory afferent activity contribute to the transition from acute to chronic pain. However, it is unlikely that a single sensory receptor is entirely responsible for persistent pain. It is more probable that extended changes to multiple receptor proteins expressed by afferent neurons support persistent pain. A-Kinase Anchoring Protein 79/150 (AKAP) is an intracellular scaffolding protein expressed in sensory neurons that spatially and temporally coordinates signaling events. Since AKAP scaffolds biochemical modifications of multiple TRP receptors linked to pain phenotypes, we probed for other ionotropic receptors that may be mediated by AKAP and contribute to persistent pain. Here, we identify a role for AKAP modulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid Receptor (AMPA-R) functionality in sensory neurons. Pharmacological manipulation of distinct AMPA-R subunits significantly reduces persistent mechanical hypersensitivity observed during hyperalgesic priming. Stimulation of both protein kinases C and A (PKC, PKA, respectively) modulate AMPA-R subunit GluR1 and GluR2 phosphorylation and surface expression in an AKAP-dependent manner in primary cultures of DRG neurons. Furthermore, AKAP knock out reduces sensitized AMPA-R responsivity in DRG neurons. Collectively, these data indicate that AKAP scaffolds AMPA-R subunit organization in DRG neurons that may contribute to the transition from acute-to-chronic pain.


Assuntos
Proteínas de Ancoragem à Quinase A , Dor Crônica , Humanos , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais
2.
eNeuro ; 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35882549

RESUMO

There are currently no Food and Drug Administration (FDA)-approved delta opioid receptor (DOR)-selective agonists, despite having fewer side effects in rodents and non-human primates compared to traditional mu opioid receptor (MOR) therapeutics (Vanderah, 2010). Targeting peripheral receptors is an attractive strategy to reduce abuse potential. However, peripheral opioid receptors do not readily respond to agonists unless primed by inflammation, which would limit their efficacy in non-inflammatory pain patients (Stein et al., 1989). It was recently identified that G protein-coupled receptor kinase 2 (GRK2) maintains DOR incompetence in non-inflamed nociceptors (Brackley et al., 2016; Brackley et al., 2017). Here, we report that paroxetine, a selective serotonin reuptake inhibitor and potent GRK2 inhibitor (Thal et al., 2012), reduces chronic GRK2 association with membrane DOR, thereby enhancing peripheral DOR-mediated analgesic competence in the absence of inflammation. Interestingly, paroxetine's effects on GRK2 in vivo are limited to peripheral tissues in the male rat. The effects of paroxetine on DOR competence are notably antagonized by GRK2 overexpression. This is the first study to suggest that paroxetine induces peripheral DOR analgesic competence through a GRK2-dependent mechanism, improving analgesic efficacy in non-inflamed tissue. Because paroxetine targets the protein that governs peripheral opioid receptor responsiveness, and does so in the absence of inflammation, we propose that paroxetine may be suitable as a co-therapy with peripherally-restrictive doses of opioids to improve analgesic efficacy in non-inflammatory pain conditions.Significance StatementOpioids that target MOR represent the gold-standard for analgesic healthcare, despite widespread abuse potential and the ongoing opioid-epidemic. Work herein uncovers the therapeutic potential of targeting peripheral DOR for analgesic utility with an FDA-approved GRK2 inhibitor paroxetine to boost efficacy and reduce side effect profiles. Analgesic pain management targeting DOR with increased efficacy through adjuvant paroxetine treatment could reduce over-reliance on MOR agonist opioids for pain relief and usher in new options for analgesia.

3.
J Neurochem ; 162(2): 156-165, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35526109

RESUMO

Inflammatory hyperalgesia represents a nociceptive phenotype that can become persistent in nature through dynamic protein modifications. However, a large gap in knowledge exists concerning how the integration of intracellular signaling molecules coordinates a persistent inflammatory phenotype. Herein, we demonstrate that Raf Kinase Anchoring Protein (RKIP) interrupts a vital canonical desensitization pathway to maintain bradykinin (BK) receptor activation in primary afferent neurons. Biochemical analyses of primary neuronal cultures indicate bradykinin-stimulated PKC phosphorylation of RKIP at Ser153. Furthermore, BK exposure increases G-protein Receptor Kinase 2 (GRK2) binding to RKIP, inhibiting pharmacological desensitization of the BK receptor. Additional studies found that molecular RKIP down-regulation increases BK receptor desensitization in real-time imaging of primary afferent neurons, identifying a key pathway integrator in the desensitization process that controls multiple GRK2-sensitive G-protein coupled receptors. Therefore, RKIP serves as an integral scaffolding protein that inhibits BK receptor desensitization.


Assuntos
Bradicinina , Receptores da Bradicinina , Bradicinina/farmacologia , Fosforilação , Transdução de Sinais , Fatores de Transcrição , Quinases raf
4.
Artigo em Inglês | MEDLINE | ID: mdl-35431176

RESUMO

OBJECTIVE: The purpose of this study was to evaluate subjective and objective outcomes in patients with temporomandibular joint (TMJ) ankylosis treated with TMJ alloplastic reconstruction (TMJR). STUDY DESIGN: All patients diagnosed with TMJ ankylosis that underwent TMJR at our institution between 2010 and 2019 were retrospectively reviewed. Patients were divided into 2 cohorts: bony and fibrous ankylosis. Subjective variables assessed were facial pain and headaches, TMJ pain, jaw function, diet, and disability. Objective variables assessed were maximum interincisal opening and lateral excursions. The Mann-Whitney test was employed to analyze subjective variables and an unpaired t-test was used to analyze the objective variables. P < .05 was considered statistically significant. RESULTS: Twenty-eight patients met the inclusion criteria (21 female, 7 male). The mean age at the time of surgery was 42 years, and the mean number of prior TMJ surgeries was 3. A total of 52 TMJRs were performed in the 28 patients, and the mean follow-up time was 46 months. All subjective variables were significantly improved, and the mean maximum interincisal opening increased from 16.9 mm to 37.25 mm. CONCLUSIONS: The results of the study demonstrate that TMJR is an effective and reliable method for the management of both fibrous and bony TMJ ankylosis.


Assuntos
Anquilose , Prótese Articular , Transtornos da Articulação Temporomandibular , Anquilose/cirurgia , Feminino , Humanos , Masculino , Estudos Retrospectivos , Articulação Temporomandibular/cirurgia , Transtornos da Articulação Temporomandibular/cirurgia
5.
J Oral Maxillofac Surg ; 80(5): 827-837, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35151639

RESUMO

PURPOSE: An extension of digital technology is to provide patient-specific hardware to reposition the first jaw in a bimaxillary case without the use of an intermediate splint. The purpose of our study was to determine if there were significant differences in maxillary repositioning using interim splints versus patient-specific guides and implants (PSIs) in executing a bimaxillary virtual surgical plan (VSP). MATERIALS AND METHODS: This is a retrospective cohort study of patients who underwent bimaxillary orthognathic surgery with interim splints or PSIs planned with VSP at our institution. The difference in maxillary positions from the VSP to the postoperative cone-beam computed tomography (CBCT) was evaluated in both groups. The primary predictor variable was the method by which the maxilla was repositioned (interim splint vs PSI). The primary outcome variable was the postoperative 3D position of the maxillary incisors and right and left first molars in the anteroposterior, transverse, and vertical dimensions. Differences in the planned and postoperative positions of the above landmarks in all three planes of space between the two groups were statistically analyzed. RESULTS: A total of 82 patients were included. 13 patients had their maxillae repositioned with an interim splint between the unoperated mandible and the mobile maxilla, and 69 patients had their maxilla repositioned using custom drill/cutting guides and a PSI. The mean difference between the planned and actual position of the maxilla in the PSI group was smaller than in the splint group. In the PSI group alone, vertical changes were accurate whether the maxilla was being superiorly or inferiorly repositioned. CONCLUSION: The use of a PSI provides more accurate maxillary repositioning during bimaxillary surgery than the use of an interim splint.


Assuntos
Procedimentos Cirúrgicos Ortognáticos , Cirurgia Assistida por Computador , Humanos , Imageamento Tridimensional , Maxila/diagnóstico por imagem , Maxila/cirurgia , Procedimentos Cirúrgicos Ortognáticos/métodos , Estudos Retrospectivos , Contenções , Cirurgia Assistida por Computador/métodos
6.
ACS Chem Neurosci ; 11(24): 4376-4386, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33174729

RESUMO

The peripheral mu-opioid receptor (MOR) has been recognized as a potential target to provide safer analgesia with reduced central side effects. Although analgesic incompetence of the peripheral MOR in the absence of inflammation was initially identified more than a decade ago, there has been very limited investigation into the underlying signaling mechanisms. Here we identify that G protein-coupled receptor kinase 2 (GRK2) constitutively interacts with the MOR in peripheral sensory neurons to suppress peripheral MOR activity. Brief exposure to bradykinin (BK) causes uncoupling of GRK2 from the MOR and subsequent restoration of MOR functionality in dorsal root ganglion (DRG) neurons. Interestingly, prolonged BK treatment induces constitutive activation of the MOR through a mechanism that involves protein kinase C (PKC) activation. After silencing Raf kinase inhibitory protein (RKIP) by RNA interference, BK-induced constitutive MOR activation is completely abrogated, which agrees with previous findings that BK activates PKC signaling to initiate GRK2 sequestration by RKIP. Furthermore, we demonstrate that constitutive, peripheral MOR activity requires GRK2 uncoupling and that the FDA-approved SSRI paroxetine promotes this state of uncoupling. Collectively, these results indicate that GRK2 tightly regulates MOR functional states and controls constitutive MOR activity in peripheral sensory neurons, supporting the potential for targeting the kinase to provide safer analgesia.


Assuntos
Analgesia , Quinase 2 de Receptor Acoplado a Proteína G , Receptores Opioides mu , Analgésicos Opioides/farmacologia , Animais , Bradicinina/farmacologia , Células Cultivadas , Gânglios Espinais , Ratos , Células Receptoras Sensoriais , Transdução de Sinais
7.
FASEB J ; 34(1): 287-302, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914619

RESUMO

Unique features of sensory neuron subtypes are manifest by their distinct physiological and pathophysiological functions. Using patch-clamp electrophysiology, Ca2+ imaging, calcitonin gene-related peptide release assay from tissues, protein biochemistry approaches, and behavioral physiology on pain models, this study demonstrates the diversity of sensory neuron pathophysiology is due in part to subtype-dependent sensitization of TRPV1 and TRPA1. Differential sensitization is influenced by distinct expression of inflammatory mediators, such as prostaglandin E2 (PGE2), bradykinin (BK), and nerve growth factor (NGF) as well as multiple kinases, including protein kinase A (PKA) and C (PKC). However, the co-expression and interaction of TRPA1 with TRPV1 proved to be the most critical for differential sensitization of sensory neurons. We identified N- and C-terminal domains on TRPV1 responsible for TRPA1-TRPV1 (A1-V1) complex formation. Ablation of A1-V1 complex with dominant-negative peptides against these domains substantially reduced the sensitization of TRPA1, as well as BK- and CFA-induced hypersensitivity. These data indicate that often occurring TRP channel complexes regulate diversity in neuronal sensitization and may provide a therapeutic target for many neuroinflammatory pain conditions.


Assuntos
Cálcio/metabolismo , Gânglios Espinais/fisiologia , Hipersensibilidade/patologia , Dor/patologia , Células Receptoras Sensoriais/fisiologia , Canal de Cátion TRPA1/fisiologia , Canais de Cátion TRPV/fisiologia , Animais , Gânglios Espinais/citologia , Hipersensibilidade/metabolismo , Masculino , Camundongos , Camundongos Knockout , Nociceptividade , Dor/metabolismo , Células Receptoras Sensoriais/citologia
8.
Mol Pharmacol ; 95(5): 463-467, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30723091

RESUMO

Opioids serve a vital role in the current analgesic array of treatment options. They are useful in acute instances involving severe pain associated with trauma, surgery, and terminal diseases such as cancer. In the past three decades, multiple receptor isoforms and conformations have been reported throughout literature. Most of these studies conducted systemic analyses of opioid receptor function, often generalizing findings from receptor systems in central nervous tissue or exogenously expressing immortalized cell lines as common mechanisms throughout physiology. However, a culmination of innovative experimental data indicates that opioid receptor systems are differentially modulated depending on their anatomic expression profile. Importantly, opioid receptors expressed in the peripheral nervous system undergo regulation uncommon to similar receptors expressed in central nervous system tissues. This distinctive characteristic begs one to question whether peripheral opioid receptors maintain anatomically unique roles, and whether they may serve an analgesic advantage in providing pain relief without promoting addiction.


Assuntos
Sistema Nervoso Periférico/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Humanos , Dor/metabolismo
9.
Pain Rep ; 3(3): e658, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29922747

RESUMO

INTRODUCTION: Chronic metabotropic glutamate receptor activation in nociceptive afferents may upregulate A-Kinase Anchoring Protein 150 (AKAP150) expression and/or function. OBJECTIVES: To quantify transcriptional changes in AKAP150 expression and/or function after long-term mGluR5 agonist exposure, and identify transcriptional elements responsible. METHODS: Dorsal root ganglia (DRG) were dissected from Sprague-Dawley rats and cultured for biochemical analysis of AKAP150 expression after prolonged mGluR5 agonist exposure. Serum response factor (SRF) expression was knocked down through siRNA in cultures to demonstrate significance to AKAP150 upregulation. Serum response factor was also knocked down in vivo through intrathecal injections of specifically targeted oligonucleotides to demonstrate significance to hyperalgesic priming behavior in persistent mechanical hypersensitivity. RESULTS: Serum response factor and AKAP150 are coexpressed in TRPV1(+) DRG neurons in intact DRG. Prolonged mGluR5 agonist exposure increases SRF-dependent transcription and AKAP150 expression in a manner sensitive to protein kinase C inhibition and SRF knock down. Serum response factor in vivo knock down reduces mechanical hyperalgesic priming. CONCLUSION: Serum response factor transcription plays an important role in transcriptional upregulation of AKAP and hyperalgesic priming behavior, and may contribute to the increased role of AKAP150 in the transition from acute to chronic pain.

10.
PLoS One ; 12(8): e0182102, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28797041

RESUMO

Blast-associated sensory and cognitive trauma sustained by military service members is an area of extensively studied research. Recent studies in our laboratory have revealed that low-level blast exposure increased expression of transient receptor potential vanilloid 1 (TRPV1) and endothelin-1 (ET-1), proteins well characterized for their role in mediating pain transmission, in the cornea. Determining the functional consequences of these alterations in protein expression is critical to understanding blast-related sensory trauma. Thus, the purpose of this study was to examine TRPV1 and ET-1 expression in ocular associated sensory tissues following primary and tertiary blast. A rodent model of blast injury was used in which anesthetized animals, unrestrained or restrained, received a single or repeat blast (73.8 ± 5.5 kPa) from a compressed air shock tube once or daily for five consecutive days, respectively. Behavioral and functional analyses were conducted to assess blast effects on nocifensive behavior and TRPV1 activity. Immunohistochemistry and Western Blot were also performed with trigeminal ganglia (TG) to determine TRPV1, ET-1 and glial fibrillary associated protein (GFAP) expression following blast. Increased TRPV1, ET-1 and GFAP were detected in the TG of animals exposed to repeat blast. Increased nocifensive responses were also observed in animals exposed to repeat, tertiary blast as compared to single blast and control. Moreover, decreased TRPV1 desensitization was observed in TG neurons exposed to repeat blast. Repeat, tertiary blast resulted in increased TRPV1, ET-1 and GFAP expression in the TG, enhanced nociception and decreased TRPV1 desensitization.


Assuntos
Traumatismos por Explosões/metabolismo , Endotelina-1/metabolismo , Neurônios/metabolismo , Canais de Cátion TRPV/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Masculino , Ratos , Ratos Long-Evans
11.
Sci Rep ; 7(1): 1842, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28500286

RESUMO

Mechanical pain serves as a base clinical symptom for many of the world's most debilitating syndromes. Ion channels expressed by peripheral sensory neurons largely contribute to mechanical hypersensitivity. Transient Receptor Potential A 1 (TRPA1) is a ligand-gated ion channel that contributes to inflammatory mechanical hypersensitivity, yet little is known as to the post-translational mechanism behind its somatosensitization. Here, we utilize biochemical, electrophysiological, and behavioral measures to demonstrate that metabotropic glutamate receptor-induced sensitization of TRPA1 nociceptors stimulates targeted modification of the receptor. Type 1 mGluR5 activation increases TRPA1 receptor agonist sensitivity in an AKA-dependent manner. As a scaffolding protein for Protein Kinases A and C (PKA and PKC, respectively), AKAP facilitates phosphorylation and sensitization of TRPA1 in ex vivo sensory neuronal preparations. Furthermore, hyperalgesic priming of mechanical hypersensitivity requires both TRPA1 and AKAP. Collectively, these results identify a novel AKAP-mediated biochemical mechanism that increases TRPA1 sensitivity in peripheral sensory neurons, and likely contributes to persistent mechanical hypersensitivity.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Canal de Cátion TRPA1/metabolismo , Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/genética , Animais , Células CHO , Cálcio/metabolismo , Cromatografia Líquida , Cricetulus , Masculino , Camundongos , Camundongos Knockout , Imagem Molecular , Fosforilação , Ratos , Receptores de Glutamato Metabotrópico/química , Canal de Cátion TRPA1/química , Canal de Cátion TRPA1/genética , Espectrometria de Massas em Tandem
12.
J Biol Chem ; 292(21): 8762-8772, 2017 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-28381559

RESUMO

µ-Opioid receptor (MOR) agonists are often used to treat severe pain but can result in adverse side effects. To circumvent systemic side effects, targeting peripheral opioid receptors is an attractive alternative treatment for severe pain. Activation of the δ-opioid receptor (DOR) produces similar analgesia with reduced side effects. However, until primed by inflammation, peripheral DOR is analgesically incompetent, raising interest in the mechanism. We recently identified a novel role for G-protein-coupled receptor kinase 2 (GRK2) that renders DOR analgesically incompetent at the plasma membrane. However, the mechanism that maintains constitutive GRK2 association with DOR is unknown. Protein kinase A (PKA) phosphorylation of GRK2 at Ser-685 targets it to the plasma membrane. Protein kinase A-anchoring protein 79/150 (AKAP), residing at the plasma membrane in neurons, scaffolds PKA to target proteins to mediate downstream signal. Therefore, we sought to determine whether GRK2-mediated DOR desensitization is directed by PKA via AKAP scaffolding. Membrane fractions from cultured rat sensory neurons following AKAP siRNA transfection and from AKAP-knock-out mice had less PKA activity, GRK2 Ser-685 phosphorylation, and GRK2 plasma membrane targeting than controls. Site-directed mutagenesis revealed that GRK2 Ser-685 phosphorylation drives the association of GRK2 with plasma membrane-associated DOR. Moreover, overexpression studies with AKAP mutants indicated that impaired AKAP-mediated PKA scaffolding significantly reduces DOR-GRK2 association at the plasma membrane and consequently increases DOR activity in sensory neurons without a priming event. These findings suggest that AKAP scaffolds PKA to increase plasma membrane targeting and phosphorylation of GRK2 to maintain DOR analgesic incompetence in peripheral sensory neurons.


Assuntos
Membrana Celular/metabolismo , Receptores Opioides delta/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Bovinos , Membrana Celular/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Masculino , Camundongos , Fosforilação/genética , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Células Receptoras Sensoriais/patologia
13.
Cell Rep ; 16(10): 2686-2698, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27568556

RESUMO

Opioids remain the standard for analgesic care; however, adverse effects of systemic treatments contraindicate long-term administration. While most clinical opioids target mu opioid receptors (MOR), those that target the delta class (DOR) also demonstrate analgesic efficacy. Furthermore, peripherally restrictive opioids represent an attractive direction for analgesia. However, opioid receptors including DOR are analgesically incompetent in the absence of inflammation. Here, we report that G protein-coupled receptor kinase 2 (GRK2) naively associates with plasma membrane DOR in peripheral sensory neurons to inhibit analgesic agonist efficacy. This interaction prevents optimal Gß subunit association with the receptor, thereby reducing DOR activity. Importantly, bradykinin stimulates GRK2 movement away from DOR and onto Raf kinase inhibitory protein (RKIP). protein kinase C (PKC)-dependent RKIP phosphorylation induces GRK2 sequestration, restoring DOR functionality in sensory neurons. Together, these results expand the known function of GRK2, identifying a non-internalizing role to maintain peripheral DOR in an analgesically incompetent state.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Receptores Opioides delta/metabolismo , Analgésicos Opioides/farmacologia , Animais , Bradicinina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Masculino , Nociceptividade/efeitos dos fármacos , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo
14.
Pain ; 156(11): 2364-2372, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26172554

RESUMO

Glutamate serves as the primary excitatory neurotransmitter in the nervous system. Previous studies have identified a role for glutamate and group I metabotropic receptors as targets for study in peripheral inflammatory pain. However, the coordination of signaling events that transpire from receptor activation to afferent neuronal sensitization has not been explored. Herein, we identify that scaffolding protein A-kinase anchoring protein 79/150 (AKAP150) coordinates increased peripheral thermal sensitivity after group I metabotropic receptor (mGluR5) activation. In both acute and persistent models of thermal somatosensory behavior, we report that mGluR5 sensitization requires AKAP150 expression. Furthermore, electrophysiological approaches designed to record afferent neuronal activity reveal that mGluR5 sensitization also requires functional AKAP150 expression. In dissociated primary afferent neurons, mGluR5 activation increases TRPV1 responses in an AKAP-dependent manner through a mechanism that induces AKAP association with TRPV1. Experimental results presented herein identify a mechanism of receptor-driven scaffolding association with ion channel targets. Importantly, this mechanism could prove significant in the search for therapeutic targets that repress episodes of acute pain from becoming chronic in nature.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Regulação da Expressão Gênica/genética , Receptores de Glutamato Metabotrópico/metabolismo , Células Receptoras Sensoriais/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Animais , Células Cultivadas , Estrenos/farmacologia , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Condução Nervosa/fisiologia , Neurotransmissores/farmacologia , Limiar da Dor/fisiologia , Nervos Periféricos/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Estimulação Física/efeitos adversos , Pirrolidinonas/farmacologia , Ratos , Células Receptoras Sensoriais/efeitos dos fármacos , Pele/inervação , Gânglio Trigeminal/citologia
15.
J Neurosci ; 35(22): 8593-603, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041925

RESUMO

Nerve growth factor (NGF) is elevated in certain chronic pain conditions and is a sufficient stimulus to cause lasting pain in humans, but the actual mechanisms underlying the persistent effects of NGF remain incompletely understood. We developed a rat model of NGF-induced persistent thermal hyperalgesia and mechanical allodynia to determine the role of transient receptor potential vanilloid 1 (TRPV1) and oxidative mechanisms in the persistent effects of NGF. Persistent thermal hypersensitivity and mechanical allodynia require de novo protein translation and are mediated by TRPV1 and oxidative mechanisms. By comparing effects after systemic (subcutaneous), spinal (intrathecal) or hindpaw (intraplantar) injections of test compounds, we determined that TRPV1 and oxidation mediate persistent thermal hypersensitivity via peripheral and spinal sites of action and mechanical allodynia via only a spinal site of action. Therefore, NGF-evoked thermal and mechanical allodynia are mediated by spatially distinct mechanisms. NGF treatment evoked sustained increases in peripheral and central TRPV1 activity, as demonstrated by increased capsaicin-evoked nocifensive responses, increased calcitonin gene-related peptide release from hindpaw skin biopsies, and increased capsaicin-evoked inward current and membrane expression of TRPV1 protein in dorsal root ganglia neurons. Finally, we showed that NGF treatment increased concentrations of linoleic and arachidonic-acid-derived oxidized TRPV1 agonists in spinal cord and skin biopsies. Furthermore, increases in oxidized TRPV1-active lipids were reduced by peripheral and spinal injections of compounds that completely blocked persistent nociception. Collectively, these data indicate that NGF evokes a persistent nociceptive state mediated by increased TRPV1 activity and oxidative mechanisms, including increased production of oxidized lipid TRPV1 agonists.


Assuntos
Fator de Crescimento Neural/farmacologia , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Estresse Oxidativo/fisiologia , Canais de Cátion TRPV/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capsaicina/farmacologia , Células Cultivadas , Cicloeximida/farmacologia , Ensaio de Imunoadsorção Enzimática , Gânglios Espinais/citologia , Hiperalgesia/etiologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Medição da Dor , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Fármacos do Sistema Sensorial/farmacologia , Pele/inervação
16.
ASN Neuro ; 7(2)2015.
Artigo em Inglês | MEDLINE | ID: mdl-25873305

RESUMO

Endothelin-1 (ET-1) and bradykinin (BK) are endogenous peptides that signal through Gαq/11-protein coupled receptors (GPCRs) to produce nociceptor sensitization and pain. Both peptides activate phospholipase C to stimulate Ca(2+) accumulation, diacylglycerol production, and protein kinase C activation and are rapidly desensitized via a G-protein receptor kinase 2-dependent mechanism. However, ET-1 produces a greater response and longer lasting nocifensive behavior than BK in multiple models, indicating a potentially divergent signaling mechanism in primary afferent sensory neurons. Using cultured sensory neurons, we demonstrate significant differences in both Ca(2+) influx and Ca(2+) release from intracellular stores following ET-1 and BK treatments. As intracellular store depletion may contribute to the regulation of other signaling cascades downstream of GPCRs, we concentrated our investigation on store-operated Ca(2+) channels. Using pharmacological approaches, we identified transient receptor potential canonical channel 3 (TRPC3) as a dominant contributor to Ca(2+) influx subsequent to ET-1 treatment. On the other hand, BK treatment stimulated Orai1 activation, with only minor input from TRPC3. Taken together, data presented here suggest that ET-1 signaling targets TRPC3, generating a prolonged Ca(2+) signal that perpetuates nocifensive responses. In contrast, Orai1 dominates as the downstream target of BK receptor activation and results in transient intracellular Ca(2+) increases and abridged nocifensive responses.


Assuntos
Bradicinina/metabolismo , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Endotelina-1/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Células Cultivadas , , Masculino , Dor Nociceptiva/metabolismo , Proteína ORAI1 , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/fisiologia , Fosfolipases Tipo C/metabolismo
17.
Prog Mol Biol Transl Sci ; 131: 31-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25744669

RESUMO

Peripheral injury precipitates the release and accumulation of extracellular molecules at the site of injury. Although these molecules exist in various forms, they activate specific receptor classes expressed on primary afferent neurons to mediate cellular and behavioral responses to both nonpainful and painful stimuli. These inflammatory mediators and subsequent receptor-mediated effects exist to warn an organism of future injury, thereby resulting in protection and rehabilitation of the wounded tissue. In this chapter, inflammatory mediators, their target receptor classes, and downstream signaling pathways are identified and discussed within the context of inflammatory hyperalgesia. Furthermore, scaffolding mechanisms that exist to support inflammatory signaling in peripheral afferent neuronal tissues specifically are identified and discussed. Together, the mediators, pathways, and scaffolding mechanisms involved in inflammatory hyperalgesia provide a unique knowledge point from which new therapeutic targets can be understood.


Assuntos
Inflamação/patologia , Dor/metabolismo , Transdução de Sinais , Animais , Humanos , Mediadores da Inflamação/metabolismo , Modelos Biológicos
18.
Neuron ; 85(4): 833-46, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25640077

RESUMO

TRPA1 and TRPV1 are crucial pain mediators, but how their interaction contributes to persistent pain is unknown. Here, we identify Tmem100 as a potentiating modulator of TRPA1-V1 complexes. Tmem100 is coexpressed and forms a complex with TRPA1 and TRPV1 in DRG neurons. Tmem100-deficient mice show a reduction in inflammatory mechanical hyperalgesia and TRPA1- but not TRPV1-mediated pain. Single-channel recording in a heterologous system reveals that Tmem100 selectively potentiates TRPA1 activity in a TRPV1-dependent manner. Mechanistically, Tmem100 weakens the association of TRPA1 and TRPV1, thereby releasing the inhibition of TRPA1 by TRPV1. A Tmem100 mutant, Tmem100-3Q, exerts the opposite effect; i.e., it enhances the association of TRPA1 and TRPV1 and strongly inhibits TRPA1. Strikingly, a cell-permeable peptide (CPP) containing the C-terminal sequence of Tmem100-3Q mimics its effect and inhibits persistent pain. Our study unveils a context-dependent modulation of the TRPA1-V1 complex, and Tmem100-3Q CPP is a promising pain therapy.


Assuntos
Proteínas de Membrana/metabolismo , Dor/metabolismo , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Células CHO , Capsaicina/toxicidade , Células Cultivadas , Cricetulus , Modelos Animais de Doenças , Estimulação Elétrica , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Dor/induzido quimicamente , Dor/patologia , Medição da Dor , Estimulação Física , Canal de Cátion TRPA1
19.
Mol Pain ; 10: 50, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25085415

RESUMO

Despite advances in understanding the signaling mechanisms involved in the development and maintenance of chronic pain, the pharmacologic treatment of chronic pain has seen little advancement. Agonists at the mu opioid receptor (MOPr) continue to be vital in the treatment of many forms of chronic pain, but side-effects limit their clinical utility and range from relatively mild, such as constipation, to major, such as addiction and dependence. Additionally, chronic activation of MOPr results in pain hypersensitivity known as opioid-induced hyperalgesia (OIH), and we have shown recently that recruitment of ß-arrestin2 to MOPr, away from transient potential vanilloid eceptor type 1 (TRPV1) in primary sensory neurons contributes to this phenomenon. The delta opioid receptor (DOPr) has become a promising target for the treatment of chronic pain, but little is known about the effects of chronic activation of DOPr on nociceptor sensitivity and OIH. Here we report that chronic activation of DOPr by the DOPr-selective agonist, SNC80, results in the sensitization of TRPV1 and behavioral signs of OIH via ß-arrestin2 recruitment to DOPr and away from TRPV1. Conversely, chronic treatment with ARM390, a DOPr-selective agonist that does not recruit ß-arrestin2, neither sensitized TRPV1 nor produced OIH. Interestingly, the effect of SNC80 to sensitize TRPV1 is species-dependent, as rats developed OIH but mice did not. Taken together, the reported data identify a novel side-effect of chronic administration of ß-arrestin2-biased DOPr agonists and highlight the importance of potential species-specific effects of DOPr agonists.


Assuntos
Arrestinas/metabolismo , Receptores Opioides mu/agonistas , Células Receptoras Sensoriais/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Animais , Benzamidas/farmacologia , Capsaicina/toxicidade , Células Cultivadas , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Lectinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Limiar da Dor/efeitos dos fármacos , Piperazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fármacos do Sistema Sensorial/toxicidade , Especificidade da Espécie , Fatores de Tempo , Gânglio Trigeminal , beta-Arrestina 2 , beta-Arrestinas
20.
PLoS One ; 9(4): e93688, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24695785

RESUMO

The transient receptor potential family V1 channel (TRPV1) is activated by multiple stimuli, including capsaicin, acid, endovanilloids, and heat (>42C). Post-translational modifications to TRPV1 result in dynamic changes to the sensitivity of receptor activation. We have previously demonstrated that ß-arrestin2 actively participates in a scaffolding mechanism to inhibit TRPV1 phosphorylation, thereby reducing TRPV1 sensitivity. In this study, we evaluated the effect of ß-arrestin2 sequestration by G-protein coupled receptors (GPCRs) on thermal and chemical activation of TRPV1. Here we report that activation of mu opioid receptor by either morphine or DAMGO results in ß-arrestin2 recruitment to mu opioid receptor in sensory neurons, while activation by herkinorin does not. Furthermore, treatment of sensory neurons with morphine or DAMGO stimulates ß-arrestin2 dissociation from TRPV1 and increased sensitivity of the receptor. Conversely, herkinorin treatment has no effect on TRPV1 sensitivity. Additional behavioral studies indicate that GPCR-driven ß-arrestin2 sequestration plays an important peripheral role in the development of thermal sensitivity. Taken together, the reported data identify a novel cross-talk mechanism between GPCRs and TRPV1 that may contribute to multiple clinical conditions.


Assuntos
Arrestinas/metabolismo , Receptores Opioides mu/agonistas , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Transferência Ressonante de Energia de Fluorescência , Furanos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Pironas/farmacologia , beta-Arrestina 2 , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...