Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Pain Res (Lausanne) ; 5: 1372942, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38721062

RESUMEN

This study investigates the impact of combining psychophysical stress, induced by forced swim (FSS), with masseter inflammation on reactive oxygen species (ROS) production in trigeminal ganglia (TG), TRPA1 upregulation in TG, and mechanical hyperalgesia. In a rat model, we demonstrate that FSS potentiates and prolongs CFA-induced ROS upregulation within TG. The ROS levels in CFA combined with FSS group surpass those in the CFA-only group on days 4 and 28 post-treatment. FSS also enhances TRPA1 upregulation in TG, with prolonged expression compared to CFA alone. Furthermore, CFA-induced mechanical hyperalgesia is significantly prolonged by FSS, persisting up to day 28. PCR array analyses reveal distinct alterations in oxidative stress genes under CFA and CFA combined with FSS conditions, suggesting an intricate regulation of ROS within TG. Notably, genes like Nox4, Hba1, Gpx3, and Duox1 exhibit significant changes, providing potential targets for managing oxidative stress and inflammatory pain. Western blot and immunohistochemistry confirm DUOX1 protein upregulation and localization in TG neurons, indicating a role in ROS generation under inflammatory and stress conditions. This study underscores the complex interplay between psychophysical stress, inflammation, and oxidative stress in the trigeminal system, offering insights into novel therapeutic targets for pain management.

2.
Pain ; 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38285538

RESUMEN

ABSTRACT: Metabolism is inextricably linked to every aspect of cellular function. In addition to energy production and biosynthesis, metabolism plays a crucial role in regulating signal transduction and gene expression. Altered metabolic states have been shown to maintain aberrant signaling and transcription, contributing to diseases like cancer, cardiovascular disease, and neurodegeneration. Metabolic gene polymorphisms and defects are also associated with chronic pain conditions, as are increased levels of nerve growth factor (NGF). However, the mechanisms by which NGF may modulate sensory neuron metabolism remain unclear. This study demonstrated that intraplantar NGF injection reprograms sensory neuron metabolism. Nerve growth factor suppressed mitochondrial pyruvate oxidation and enhanced lactate extrusion, requiring 24 hours to increase lactate dehydrogenase A and pyruvate dehydrogenase kinase 1 (PDHK1) expression. Inhibiting these metabolic enzymes reversed NGF-mediated effects. Remarkably, directly disrupting mitochondrial pyruvate oxidation induced severe, persistent allodynia, implicating this metabolic dysfunction in chronic pain. Nanopore long-read sequencing of poly(A) mRNA uncovered extensive transcriptomic changes upon metabolic disruption, including altered gene expression, splicing, and poly(A) tail lengths. By linking metabolic disturbance of dorsal root ganglia to transcriptome reprogramming, this study enhances our understanding of the mechanisms underlying persistent nociceptive sensitization. These findings imply that impaired mitochondrial pyruvate oxidation may drive chronic pain, possibly by impacting transcriptomic regulation. Exploring these metabolite-driven mechanisms further might reveal novel therapeutic targets for intractable pain.

3.
Mol Pain ; 15: 1744806919850043, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31041875

RESUMEN

Chemotherapy-induced painful peripheral neuropathy is a significant clinical problem that is associated with widely used chemotherapeutics. Unfortunately, the molecular mechanisms by which chemotherapy-induced painful peripheral neuropathy develops have remained elusive. The proteasome inhibitor, bortezomib, has been shown to induce aerobic glycolysis in sensory neurons. This altered metabolic phenotype leads to the extrusion of metabolites which sensitize primary afferents and cause pain. Hypoxia-inducible factor alpha is a transcription factor that is known to reprogram cellular metabolism. Furthermore, hypoxia-inducible factor 1 alpha protein is constantly synthesized and undergoes proteasomal degradation in normal conditions. However, metabolic stress or hypoxia stabilizes the expression of hypoxia-inducible factor 1 alpha leading to the transcription of genes that reprogram cellular metabolism. This study demonstrates that treatment of mice with bortezomib stabilizes the expression of hypoxia-inducible factor 1 alpha. Moreover, knockdown of hypoxia-inducible factor 1 alpha, inhibition of hypoxia-inducible factor 1 alpha binding to its response element, or limiting its translation by using metformin prevent the development of bortezomib-induced neuropathic pain. Strikingly, the blockade of hypoxia-inducible factor 1 alpha expression does not attenuate mechanical allodynia in mice with existing bortezomib-induced neuropathic pain. These results establish the stabilization of hypoxia-inducible factor 1 alpha expression as the molecular mechanism by which bortezomib initiates chemotherapy-induced painful peripheral neuropathy. Crucially, these findings reveal that the initiation and maintenance of bortezomib-induced neuropathic pain are regulated by distinct mechanisms.


Asunto(s)
Antineoplásicos/efectos adversos , Bortezomib/efectos adversos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metformina/efectos adversos , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Adenilato Quinasa/metabolismo , Animales , Calcio/metabolismo , Equinomicina/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Hiperalgesia/patología , Masculino , Ratones Endogámicos ICR , Modelos Biológicos , Neuralgia/complicaciones , Enfermedades del Sistema Nervioso Periférico/complicaciones , Unión Proteica , Elementos de Respuesta/genética
4.
Mol Pain ; 15: 1744806919837429, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30810076

RESUMEN

Chemotherapy-induced painful peripheral neuropathy (CIPN) is the most common toxicity associated with widely used chemotherapeutics. CIPN is the major cause of dose reduction or discontinuation of otherwise life-saving treatment. Unfortunately, CIPN can persist in cancer survivors, which adversely affects their quality of life. Moreover, available treatments are vastly inadequate, warranting a better understanding of the biochemical and metabolic mechanisms that occur in response to chemotherapeutics which would be critical for the development of novel therapies for CIPN. Using extracellular flux analysis, this study demonstrated that the proteasome inhibitor, bortezomib, enhanced glycolysis while suppressing oxidative phosphorylation in the sensory neurons of mice. This metabolic phenotype is known as aerobic glycolysis. Bortezomib upregulated lactate dehydrogenase A and pyruvate dehydrogenase kinase 1, which consequently enhanced the production of lactate and repressed pyruvate oxidation, respectively. Moreover, lactate dehydrogenase A- and pyruvate dehydrogenase kinase 1-driven aerobic glycolysis was associated with increased extracellular acidification, augmented calcium responses, and pain in bortezomib-induced CIPN. Remarkably, pharmacological blockade and in vivo knockdown of lactate dehydrogenase A or pyruvate dehydrogenase kinase 1 reversed the metabolic phenotype, attenuated calcium responses, and alleviated pain induced by bortezomib. Collectively, these results elucidate the mechanisms by which bortezomib induces aerobic glycolysis. Moreover, these findings establish aerobic glycolysis as a metabolic phenotype that underpins bortezomib-induced CIPN.


Asunto(s)
Antineoplásicos/efectos adversos , Bortezomib/efectos adversos , Glucólisis/efectos de los fármacos , Dolor/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Animales , Western Blotting , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Dolor/etiología , Enfermedades del Sistema Nervioso Periférico/etiología , Calidad de Vida
5.
J Neurosci ; 37(31): 7481-7499, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28674170

RESUMEN

Injury-induced sensitization of nociceptors contributes to pain states and the development of chronic pain. Inhibiting activity-dependent mRNA translation through mechanistic target of rapamycin and mitogen-activated protein kinase (MAPK) pathways blocks the development of nociceptor sensitization. These pathways convergently signal to the eukaryotic translation initiation factor (eIF) 4F complex to regulate the sensitization of nociceptors, but the details of this process are ill defined. Here we investigated the hypothesis that phosphorylation of the 5' cap-binding protein eIF4E by its specific kinase MAPK interacting kinases (MNKs) 1/2 is a key factor in nociceptor sensitization and the development of chronic pain. Phosphorylation of ser209 on eIF4E regulates the translation of a subset of mRNAs. We show that pronociceptive and inflammatory factors, such as nerve growth factor (NGF), interleukin-6 (IL-6), and carrageenan, produce decreased mechanical and thermal hypersensitivity, decreased affective pain behaviors, and strongly reduced hyperalgesic priming in mice lacking eIF4E phosphorylation (eIF4ES209A ). Tests were done in both sexes, and no sex differences were found. Moreover, in patch-clamp electrophysiology and Ca2+ imaging experiments on dorsal root ganglion neurons, NGF- and IL-6-induced increases in excitability were attenuated in neurons from eIF4ES209A mice. These effects were recapitulated in Mnk1/2-/- mice and with the MNK1/2 inhibitor cercosporamide. We also find that cold hypersensitivity induced by peripheral nerve injury is reduced in eIF4ES209A and Mnk1/2-/- mice and following cercosporamide treatment. Our findings demonstrate that the MNK1/2-eIF4E signaling axis is an important contributing factor to mechanisms of nociceptor plasticity and the development of chronic pain.SIGNIFICANCE STATEMENT Chronic pain is a debilitating disease affecting approximately one in three Americans. Chronic pain is thought to be driven by changes in the excitability of peripheral nociceptive neurons, but the precise mechanisms controlling these changes are not elucidated. Emerging evidence demonstrates that mRNA translation regulation pathways are key factors in changes in nociceptor excitability. Our work demonstrates that a single phosphorylation site on the 5' cap-binding protein eIF4E is a critical mechanism for changes in nociceptor excitability that drive the development of chronic pain. We reveal a new mechanistic target for the development of a chronic pain state and propose that targeting the upstream kinase, MAPK interacting kinase 1/2, could be used as a therapeutic approach for chronic pain.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Dolor Crónico/fisiopatología , Factor 4E Eucariótico de Iniciación/metabolismo , Ganglios Espinales/fisiopatología , Hiperalgesia/fisiopatología , Plasticidad Neuronal , Nocicepción , Animales , Dolor Crónico/etiología , ATPasas Transportadoras de Cobre , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor Nociceptivo/etiología , Dolor Nociceptivo/fisiopatología , Células Receptoras Sensoriales/metabolismo , Transducción de Señal
6.
Brain Res Bull ; 125: 211-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27450703

RESUMEN

Activation of opioid and cannabinoid receptors expressed in nociceptors induces effective antihyperalgesia. In this study, we examined whether combinations of opioid and cannabinoid receptor agonists directed at the injured site would enhance therapeutic effectiveness. Behavioral pharmacology experiments were performed to compare the effects of DAMGO, a selective agonist for µ-opioid receptor (MOR), ACPA, a specific agonist for CB1, and combinations of DAMGO and ACPA in attenuating complete Freund's adjuvant (CFA)-induced mechanical hyperalgesia in the rat hindpaw. DAMGO (1µg-1mg) or ACPA (1µg-2mg) was administered into the inflamed paw when mechanical hyperalgesia was fully developed. When administered individually, DAMGO and ACPA dose-dependently reversed the mechanical hyperalgesia. DAMGO displayed a lower ED50 value (57.4±2.49µg) than ACPA (111.6±2.18µg), but ACPA produced longer lasting antihyperalgesic effects. Combinations of DAMGO and ACPA also dose-dependently attenuated mechanical hyperalgesia, but the antihyperalgesic effects were partial and transient even at high doses. Using isobolographic analysis, we determined that combined treatment with DAMGO and ACPA produced antagonistic effects with the observed ED50 of 128.4±2.28µg. Our findings showed that MOR and CB1 agonists directed at the inflamed site effectively attenuate mechanical hyperalgesia when administered individually, but exert opposing effects when administered together. The antagonistic interactions between the two classes of drugs at the inflamed site suggest distinct mechanisms unique to peripheral nociceptors or inflamed tissue, and therefore require further studies to investigate whether the therapeutic utility of the combined drug treatments in chronic pain conditions can be optimized.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Agonistas de Receptores de Cannabinoides/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inflamación/complicaciones , Análisis de Varianza , Animales , Ácidos Araquidónicos/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Encefalina Ala(2)-MeFe(4)-Gli(5)/uso terapéutico , Adyuvante de Freund/toxicidad , Inflamación/inducido químicamente , Masculino , Ratas , Ratas Sprague-Dawley
7.
Prog Mol Biol Transl Sci ; 131: 185-213, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25744674

RESUMEN

Pain is a crucial physiological response to injury and pathologies. The development and maintenance of pain requires the expression of novel genes. The expression of such genes occurs in highly regulated and orchestrated manner where protein translation provides an exquisite temporal and spatial fidelity within the axons and dendrites of neurons. Signaling pathways that regulate local translation are activated by cytokines, neurotrophic factors, or neurotransmitters, which are released either due to tissue damage or neuronal activity. In recent years, the ERK and mTOR pathways have been demonstrated to be central in regulating local translation in neurons of both the peripheral and central nervous systems in diverse models of chronic pain. The ERK and mTOR pathways converge onto the cap-dependent translational machinery that regulates genes essential for the development of nociceptive sensitization. Moreover, inhibition of these pathways has proved to be effective in normalizing the biochemical changes and the associated pain in various preclinical models.


Asunto(s)
Dolor Crónico/metabolismo , Biosíntesis de Proteínas , Animales , Axones/patología , Humanos , Modelos Biológicos , Transporte de ARN , Cicatrización de Heridas
8.
Pain ; 156(7): 1247-1264, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25782368

RESUMEN

Targeting proteins within the N-type voltage-gated calcium channel (CaV2.2) complex has proven to be an effective strategy for developing novel pain therapeutics. We describe a novel peptide aptamer derived from the collapsin response mediator protein 2 (CRMP2), a CaV2.2-regulatory protein. Addition of a 14-carbon myristate group to the peptide (myr-tat-CBD3) tethered it to the membrane of primary sensory neurons near surface CaV2.2. Pull-down studies demonstrated that myr-tat-CBD3 peptide interfered with the CRMP2-CaV2.2 interaction. Quantitative confocal immunofluorescence revealed a pronounced reduction of CaV2.2 trafficking after myr-tat-CBD3 treatment and increased efficiency in disrupting CRMP2-CaV2.2 colocalization compared with peptide tat-CBD3. Consequently, myr-tat-CBD3 inhibited depolarization-induced calcium influx in sensory neurons. Voltage clamp electrophysiology experiments revealed a reduction of Ca, but not Na, currents in sensory neurons after myr-tat-CBD3 exposure. Current clamp electrophysiology experiments demonstrated a reduction in excitability of small-diameter dorsal root ganglion neurons after exposure to myr-tat-CBD3. Myr-tat-CBD3 was effective in significantly attenuating carrageenan-induced thermal hypersensitivity and reversing thermal hypersensitivity induced by a surgical incision of the plantar surface of the rat hind paw, a model of postoperative pain. These effects are compared with those of tat-CBD3-the nonmyristoylated tat-conjugated CRMP2 peptide as well as scrambled versions of CBD3 and CBD3-lacking control peptides. Our results demonstrate that the myristoyl tag enhances intracellular delivery and local concentration of the CRMP2 peptide aptamer near membrane-delimited calcium channels resulting in pronounced interference with the calcium channel complex, superior suppression of calcium influx, and better antinociceptive potential.


Asunto(s)
Aptámeros de Péptidos/metabolismo , Canales de Calcio Tipo N/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/metabolismo , Transporte de Proteínas/fisiología , Secuencia de Aminoácidos , Animales , Aptámeros de Péptidos/genética , Aptámeros de Péptidos/uso terapéutico , Células Cultivadas , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/uso terapéutico , Dolor Postoperatorio/genética , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
9.
Front Cell Neurosci ; 8: 196, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25104922

RESUMEN

Activity-dependent neurite outgrowth is a highly complex, regulated process with important implications for neuronal circuit remodeling in development as well as in seizure-induced sprouting in epilepsy. Recent work has linked outgrowth to collapsin response mediator protein 2 (CRMP2), an intracellular phosphoprotein originally identified as axon guidance and growth cone collapse protein. The neurite outgrowth promoting function of CRMP2 is regulated by its phosphorylation state. In this study, depolarization (potassium chloride)-driven activity increased the level of active CRMP2 by decreasing its phosphorylation by GSK3ß via a reduction in priming by Cdk5. To determine the contribution of CRMP2 in activity-driven neurite outgrowth, we screened a limited set of compounds for their ability to reduce neurite outgrowth but not modify voltage-gated sodium channel (VGSC) biophysical properties. This led to the identification of (S)-lacosamide ((S)-LCM), a stereoisomer of the clinically used antiepileptic drug (R)-LCM (Vimpat®), as a novel tool for preferentially targeting CRMP2-mediated neurite outgrowth. Whereas (S)-LCM was ineffective in targeting VGSCs, the presumptive pharmacological targets of (R)-LCM, (S)-LCM was more efficient than (R)-LCM in subverting neurite outgrowth. Biomolecular interaction analyses revealed that (S)-LCM bound to wildtype CRMP2 with low micromolar affinity, similar to (R)-LCM. Through the use of this novel tool, the activity-dependent increase in neurite outgrowth observed following depolarization was characterized to be reliant on CRMP2 function. Knockdown of CRMP2 by siRNA in cortical neurons resulted in reduced CRMP2-dependent neurite outgrowth; incubation with (S)-LCM phenocopied this effect. Other CRMP2-mediated processes were unaffected. (S)-LCM subverted neurite outgrowth not by affecting the canonical CRMP2-tubulin association but rather by impairing the ability of CRMP2 to promote tubulin polymerization, events that are perfunctory for neurite outgrowth. Taken together, these results suggest that changes in the phosphorylation state of CRMP2 are a major contributing factor in activity-dependent regulation of neurite outgrowth.

10.
Mol Pain ; 10: 45, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24993495

RESUMEN

Transcriptional regulation of genes by cyclic AMP response element binding protein (CREB) is essential for the maintenance of long-term memory. Moreover, retrograde axonal trafficking of CREB in response to nerve growth factor (NGF) is critical for the survival of developing primary sensory neurons. We have previously demonstrated that hindpaw injection of interleukin-6 (IL-6) induces mechanical hypersensitivity and hyperalgesic priming that is prevented by the local injection of protein synthesis inhibitors. However, proteins that are locally synthesized that might lead to this effect have not been identified. We hypothesized that retrograde axonal trafficking of nascently synthesized CREB might link local, activity-dependent translation to nociceptive plasticity. To test this hypothesis, we determined if IL-6 enhances the expression of CREB and if it subsequently undergoes retrograde axonal transport. IL-6 treatment of sensory neurons in vitro caused an increase in CREB protein and in vivo treatment evoked an increase in CREB in the sciatic nerve consistent with retrograde transport. Importantly, co-injection of IL-6 with the methionine analogue azido-homoalanine (AHA), to assess nascently synthesized proteins, revealed an increase in CREB containing AHA in the sciatic nerve 2 hrs post injection, indicating retrograde transport of nascently synthesized CREB. Behaviorally, blockade of retrograde transport by disruption of microtubules or inhibition of dynein or intrathecal injection of cAMP response element (CRE) consensus sequence DNA oligonucleotides, which act as decoys for CREB DNA binding, prevented the development of IL-6-induced mechanical hypersensitivity and hyperalgesic priming. Consistent with previous studies in inflammatory models, intraplantar IL-6 enhanced the expression of BDNF in dorsal root ganglion (DRG). This effect was blocked by inhibition of retrograde axonal transport and by intrathecal CRE oligonucleotides. Collectively, these findings point to a novel mechanism of axonal translation and retrograde trafficking linking locally-generated signals to long-term nociceptive sensitization.


Asunto(s)
Transporte Axonal/efectos de los fármacos , Proteína de Unión a CREB/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/farmacología , Dolor Nociceptivo/inducido químicamente , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Transporte Axonal/fisiología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Colchicina/farmacología , Modelos Animales de Enfermedad , Ganglios Espinales/patología , Interleucina-6/toxicidad , Masculino , Ratones , Ratones Endogámicos ICR , Dolor Nociceptivo/patología , Nocodazol/farmacología , Transporte de Proteínas/efectos de los fármacos , Quinazolinonas/farmacología , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Células Receptoras Sensoriales/metabolismo , Moduladores de Tubulina/farmacología
11.
Pain ; 155(7): 1238-1244, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657451

RESUMEN

Nociceptive signaling from the meninges is proposed to contribute to many forms of headache. However, the events within the meninges that drive afferent activity are not clear. Meningeal fibroblasts are traditionally thought to produce extracellular proteins that constitute the meninges but not to contribute to headache. The purpose of these studies was to determine whether dural fibroblasts release factors that activate/sensitize dural afferents and produce headache-like behavior in rats. Dura mater was removed from male rats and dural fibroblasts were cultured. Fibroblast cultures were stimulated with vehicle or lipopolysaccharide (LPS), washed, and conditioned media was collected. Fibroblast media conditioned with vehicle or LPS was applied to retrogradely labeled rat dural trigeminal ganglion neurons in vitro. Patch-clamp electrophysiology was performed to determine whether conditioned media activated/sensitized dural afferents. A preclinical behavioral model was used where conditioned media was applied directly to the rat dura to determine the presence of cutaneous facial and hind-paw allodynia. Conditioned media was also tested for interleukin-6 (IL-6) content using an enzyme-linked immunosorbent assay. Application of LPS-conditioned fibroblast media to dural afferents produced a significant increase in action potential firing as well as cutaneous facial and hind-paw allodynia when this media was applied to the dura. Finally, stimulation of cultured fibroblasts with LPS increased IL-6 levels in the media. These findings demonstrate that fibroblasts stimulated with LPS release factors capable of activating/sensitizing dural afferents. Further, they suggest that fibroblasts play a potential role in the pathophysiology of headache.


Asunto(s)
Duramadre/metabolismo , Fibroblastos/metabolismo , Cefalea , Hiperalgesia/metabolismo , Neuronas Aferentes/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Conducta Animal , Células Cultivadas , Duramadre/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
12.
Neurosci Lett ; 563: 169-74, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24080374

RESUMEN

Processing (P) bodies are RNA granules that comprise key cellular sites for the metabolism of mRNAs. In certain cells, including neurons, these RNA granules may also play an important role in storage of mRNAs in a translationally dormant state. Utilizing nerve growth factor (NGF) and interleukin 6 (IL6), which stimulate cap-dependent translation in sensory neurons, and adenosine monophosphate activated protein kinase (AMPK) activators, which inhibit cap-dependent translation, we have tested the hypothesis that cap-dependent translation is linked to P body formation in mammalian sensory neurons. Treatment with NGF and IL6 decreases, whereas metformin increases biochemical association of the P body marker and translational repressor/decapping activator Rck/p54/dhh1 with the 5'-mRNA-cap suggesting an ordered assembly of P bodies. Likewise, diverse AMPK activators enhance P body formation while NGF and IL6 decrease P bodies in sensory neurons. This bidirectional P body plasticity readily occurs in the axonal compartment of these neurons. These studies indicate that P body formation is intricately linked to cap-dependent translation in mammalian sensory neurons suggesting an important role for these organelles in the regulation of mRNA metabolism in the adult PNS.


Asunto(s)
Estructuras Citoplasmáticas/metabolismo , Factor 4F Eucariótico de Iniciación/metabolismo , ARN/metabolismo , Células Receptoras Sensoriales/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Axones/metabolismo , Células Cultivadas , Activadores de Enzimas/farmacología , Interleucina-6/farmacología , Masculino , Ratones Endogámicos ICR , Factor de Crecimiento Nervioso/farmacología , Caperuzas de ARN/metabolismo , Ganglio del Trigémino/citología
13.
Reproduction ; 146(2): R73-80, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23690629

RESUMEN

Protein Z (PZ) is a vitamin K-dependent factor characterized by its homology to other vitamin K-dependent factors (factors VII, IX, and X, protein C and protein S), but lacks any enzymatic activity. Instead, PZ acts as a cofactor for the inhibition of factor Xa through the serpin PZ-dependent protease inhibitor (ZPI). PZ deficiency is associated with a procoagulant state, highlighted by excessive FXa secretion and thrombin production, and is linked with several thrombotic disorders, including arterial vascular and venous thromboembolic diseases. A role for the PZ-ZPI complex in the regulation of physiological pregnancy has been demonstrated, highlighted by the progressive elevation in PZ levels in the first trimester of gestation, which then steadily decline toward delivery. An association between altered plasma PZ concentrations and adverse pregnancy outcomes (recurrent miscarriage, stillbirth, preeclampsia, intrauterine growth restriction, and placental abruption) has been reported. The mechanism by which PZ deficiency leads to adverse pregnancy outcomes is not clear, but it is multifactorial. It may be attributed to the anti-PZ IgG and IgM autoantibodies, which apparently act independently of classical antiphospholipid antibodies (lupus anticoagulant, anticardiolipin, and anti-ß2-glycoprotein I antibodies). PZ deficiency has also been reported to be constitutional, and a number of variants in the PROZ (PZ) gene and SERPINA10 (ZPI) gene are linked with specific adverse pregnancy complications. This review summarizes the relationship between adverse pregnancy outcomes and acquired and constitutional PZ-ZPI deficiency, in order to understand whether or not PZ deficiency could be considered as a risk factor for poor pregnancy outcomes.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Mantenimiento del Embarazo , Autoanticuerpos/análisis , Coagulación Sanguínea , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/genética , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Femenino , Variación Genética , Humanos , Conformación Molecular , Embarazo , Complicaciones Hematológicas del Embarazo/sangre , Complicaciones Hematológicas del Embarazo/genética , Complicaciones Hematológicas del Embarazo/inmunología , Complicaciones Hematológicas del Embarazo/metabolismo , Serpinas/sangre , Serpinas/deficiencia , Serpinas/genética , Serpinas/metabolismo
14.
Pain ; 154(7): 1080-91, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23607966

RESUMEN

Mammalian target of rapamycin complex 1 (mTORC1) inhibitors are extensively used as immunosuppressants to prevent transplant rejection and in treatment of certain cancers. In patients, chronic treatment with rapamycin or its analogues (rapalogues) has been reported to lead to sensory hypersensitivity and pain conditions via an unknown mechanism. Here, we show that pharmacological or genetic inhibition of mTORC1 activates the extracellular signal-regulated kinase (ERK) pathway in sensory neurons via suppression of S6K1 to insulin receptor substrate 1 negative feedback loop. As a result, increased ERK activity induces sensory neuron sensitization, mechanical hypersensitivity, and spontaneous pain. The clinically available adenosine monophosphate-activated protein kinase activator, metformin, which is an antidiabetic drug, prevents rapamycin-induced ERK activation and the development of mechanical hypersensitivity and spontaneous pain. Taken together, our findings demonstrate that activation of the ERK pathway in sensory neurons as a consequence of mTORC1 inhibition leads to the development of pain. Importantly, this effect is abolished by co-treatment with metformin, thus providing a potential treatment option for rapalogue-evoked pain. Our findings highlight the physiological relevance of feedback signaling through mTORC1 inhibition and have important implications for development of pain therapeutics that target the mTOR pathway.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Animales , Regulación hacia Abajo , Activación Enzimática , Retroalimentación Fisiológica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Dolor/inducido químicamente , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos
15.
Mol Pain ; 9: 12, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23510079

RESUMEN

BACKGROUND: Chronic pain is an important medical problem affecting hundreds of millions of people worldwide. Mechanisms underlying the maintenance of chronic pain states are poorly understood but the elucidation of such mechanisms have the potential to reveal novel therapeutics capable of reversing a chronic pain state. We have recently shown that the maintenance of a chronic pain state is dependent on an atypical PKC, PKMζ, but the mechanisms involved in controlling PKMζ in chronic pain are completely unknown. Here we have tested the hypothesis that brain derived neurotrophic factor (BDNF) regulates PKMζ, and possibly other aPKCs, to maintain a centralized chronic pain state. RESULTS: We first demonstrate that although other kinases play a role in the initiation of persistent nociceptive sensitization, they are not involved in the maintenance of this chronic pain state indicating that a ZIP-reversible process is responsible for the maintenance of persistent sensitization. We further show that BDNF plays a critical role in initiating and maintaining persistent nociceptive sensitization and that this occurs via a ZIP-reversible process. Moreover, at spinal synapses, BDNF controls PKMζ and PKCλ nascent synthesis via mTORC1 and BDNF enhances PKMζ phosphorylaton. Finally, we show that BDNF signaling to PKMζ and PKCλ is conserved across CNS synapses demonstrating molecular links between pain and memory mechanisms. CONCLUSIONS: Hence, BDNF is a key regulator of aPKC synthesis and phosphorylation and an essential mediator of the maintenance of a centralized chronic pain state. These findings point to BDNF regulation of aPKC as a potential therapeutic target for the permanent reversal of a chronic pain state.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Dolor Crónico/enzimología , Proteína Quinasa C/metabolismo , Sinapsis/enzimología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Corteza Cerebral/patología , Dolor Crónico/patología , Factor 4F Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/enzimología , Biosíntesis de Proteínas/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Transporte de Proteínas/efectos de los fármacos , Sinapsis/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
16.
Mol Pain ; 9: 14, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23531341

RESUMEN

BACKGROUND: Peripheral nerve injury (PNI) results in a fundamental reorganization of the translational machinery in the injured peripheral nerve such that protein synthesis is increased in a manner linked to enhanced mTOR and ERK activity. We have shown that metformin treatment, which activates adenosine monophosphate-activated protein kinase (AMPK), reverses tactile allodynia and enhanced translation following PNI. To gain a better understanding of how PNI changes the proteome of the sciatic nerve and ascertain how metformin treatment may cause further change, we conducted a range of unbiased proteomic studies followed by biochemical experiments to confirm key results. RESULTS: We used multidimensional protein identification technology (MUDPIT) on sciatic nerve samples taken from rats with sham surgery, spinal nerve ligation (SNL) surgery or SNL + 200 mg/kg metformin treatment. MUDPIT analysis on these complex samples yielded a wide variety of proteins that were sorted according to their peptide counts in SNL and SNL + metformin compared to sham. These proteins were then submitted to functional annotation analysis to identify potential functional networks altered by SNL and SNL + metformin treatment. Additionally, we used click-chemistry-based labeling and purification of nascently synthesized proteins followed by MUDPIT to further identify peptides that were synthesized within the injured nerve. With these methods, we identified apolipoprotein E (ApoE) as a protein profoundly increased by PNI and further increased by PNI and metformin. This result was confirmed by Western Blot of samples from SNL rats and spared nerve injury (SNI) mice. Furthermore, we show that 7-day treatment with metformin in naïve mice leads to an increase in ApoE expression in the sciatic nerve. CONCLUSIONS: These proteomic findings support the hypothesis that PNI leads to a fundamental reorganization of gene expression within the injured nerve. Our data identify a key association of ApoE with PNI that is regulated by metformin treatment. We conclude from the known functions of ApoE in the nervous system that ApoE may be an intrinsic factor linked to nerve regeneration after PNI, an effect that is further enhanced by metformin treatment.


Asunto(s)
Apolipoproteínas E/metabolismo , Metformina/farmacología , Anotación de Secuencia Molecular , Proteómica , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Algoritmos , Animales , Masculino , Ratones , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología
17.
Pain ; 153(6): 1263-1273, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22482911

RESUMEN

Injuries can induce adaptations in pain processing that result in amplification of signaling. One mechanism may be analogous to long-term potentiation and involve the atypical protein kinase C, PKMζ. The possible contribution of PKMζ-dependent and independent amplification mechanisms to experimental neuropathic pain was explored in rats with spinal nerve ligation (SNL) injury. SNL increased p-PKMζ in the rostral anterior cingulate cortex (rACC), a site that mediates, in part, the unpleasant aspects of pain. Inhibition of PKMζ within the rACC by a single administration of ζ-pseudosubstrate inhibitory peptide (ZIP) reversed SNL-induced aversiveness within 24 hours, whereas N-methyl-d-aspartate receptor blockade with MK-801 had no effects. The SNL-induced aversive state (reflecting "spontaneous" pain), was re-established in a time-dependent manner, with full recovery observed 7 days post-ZIP administration. Neither rACC ZIP nor MK-801 altered evoked responses. In contrast, spinal ZIP or MK-801, but not scrambled peptide, transiently reversed evoked hypersensitivity, but had no effect on nerve injury-induced spontaneous pain. PKMζ phosphorylation was not altered by SNL in the spinal dorsal horn. These data suggest that amplification mechanisms contribute to different aspects of neuropathic pain at different levels of the neuraxis. Thus, PKMζ-dependent amplification contributes to nerve injury-induced aversiveness within the rACC. Moreover, unlike mechanisms maintaining memory, the consequences of PKMζ inhibition within the rACC are not permanent in neuropathic pain, possibly reflecting the re-establishment of amplification mechanisms by ongoing activity of injured nerves. In the spinal cord, however, both PKMζ-dependent and independent mechanisms contribute to amplification of evoked responses, but apparently not spontaneous pain.


Asunto(s)
Giro del Cíngulo/enzimología , Neuralgia/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Médula Espinal/enzimología , Animales , Maleato de Dizocilpina/farmacología , Masculino , Neuralgia/fisiopatología , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Nervios Espinales/enzimología , Nervios Espinales/lesiones
18.
Mol Pain ; 8: 5, 2012 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-22269797

RESUMEN

BACKGROUND: Despite advances in our understanding of basic mechanisms driving post-surgical pain, treating incision-induced pain remains a major clinical challenge. Moreover, surgery has been implicated as a major cause of chronic pain conditions. Hence, more efficacious treatments are needed to inhibit incision-induced pain and prevent the transition to chronic pain following surgery. We reasoned that activators of AMP-activated protein kinase (AMPK) may represent a novel treatment avenue for the local treatment of incision-induced pain because AMPK activators inhibit ERK and mTOR signaling, two important pathways involved in the sensitization of peripheral nociceptors. RESULTS: To test this hypothesis we used a potent and efficacious activator of AMPK, resveratrol. Our results demonstrate that resveratrol profoundly inhibits ERK and mTOR signaling in sensory neurons in a time- and concentration-dependent fashion and that these effects are mediated by AMPK activation and independent of sirtuin activity. Interleukin-6 (IL-6) is thought to play an important role in incision-induced pain and resveratrol potently inhibited IL-6-mediated signaling to ERK in sensory neurons and blocked IL-6-mediated allodynia in vivo through a local mechanism of action. Using a model of incision-induced allodynia in mice, we further demonstrate that local injection of resveratrol around the surgical wound strongly attenuates incision-induced allodynia. Intraplantar IL-6 injection and plantar incision induces persistent nociceptive sensitization to PGE2 injection into the affected paw after the resolution of allodynia to the initial stimulus. We further show that resveratrol treatment at the time of IL-6 injection or plantar incision completely blocks the development of persistent nociceptive sensitization consistent with the blockade of a transition to a chronic pain state by resveratrol treatment. CONCLUSIONS: These results highlight the importance of signaling to translation control in peripheral sensitization of nociceptors and provide further evidence for activation of AMPK as a novel treatment avenue for acute and chronic pain states.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dolor Agudo/tratamiento farmacológico , Dolor Crónico/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Receptoras Sensoriales/enzimología , Estilbenos/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Dolor Agudo/enzimología , Dolor Agudo/patología , Animales , Dolor Crónico/enzimología , Dolor Crónico/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Factor 4F Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Hiperalgesia/patología , Interleucina-6/administración & dosificación , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Dolor Postoperatorio/complicaciones , Dolor Postoperatorio/patología , Biosíntesis de Proteínas/efectos de los fármacos , Resveratrol , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Estilbenos/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factores de Tiempo , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología
19.
Mol Pain ; 8: 6, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22273495

RESUMEN

BACKGROUND: Migraine headache is one of the most common neurological disorders, but the pathophysiology contributing to migraine is poorly understood. Intracranial interleukin-6 (IL-6) levels have been shown to be elevated during migraine attacks, suggesting that this cytokine may facilitate pain signaling from the meninges and contribute to the development of headache. METHODS: Cutaneous allodynia was measured in rats following stimulation of the dura with IL-6 alone or in combination with the MEK inhibitor, U0126. The number of action potentials and latency to the first action potential peak in response to a ramp current stimulus as well as current threshold were measured in retrogradely-labeled dural afferents using patch-clamp electrophysiology. These recordings were performed in the presence of IL-6 alone or in combination with U0126. Association between ERK1 and Nav1.7 following IL-6 treatment was also measured by co-immunoprecipitation. RESULTS: Here we report that in awake animals, direct application of IL-6 to the dura produced dose-dependent facial and hindpaw allodynia. The MEK inhibitor U0126 blocked IL-6-induced allodynia indicating that IL-6 produced this behavioral effect through the MAP kinase pathway. In trigeminal neurons retrogradely labeled from the dura, IL-6 application decreased the current threshold for action potential firing. In response to a ramp current stimulus, cells treated with IL-6 showed an increase in the numbers of action potentials and a decrease in latency to the first spike, an effect consistent with phosphorylation of the sodium channel Nav1.7. Pretreatment with U0126 reversed hyperexcitability following IL-6 treatment. Moreover, co-immunoprecipitation experiments demonstrated an increased association between ERK1 and Nav1.7 following IL-6 treatment. CONCLUSIONS: Our results indicate that IL-6 enhances the excitability of dural afferents likely via ERK-mediated modulation of Nav1.7 and these responses contribute to migraine-related pain behavior in vivo. These data provide a cellular mechanism by which IL-6 in the meninges causes sensitization of dural afferents therefore contributing to the pathogenesis of migraine headache.


Asunto(s)
Conducta Animal/efectos de los fármacos , Duramadre/efectos de los fármacos , Duramadre/patología , Interleucina-6/farmacología , Meninges/fisiopatología , Trastornos Migrañosos/fisiopatología , Neuronas Aferentes/patología , Potenciales de Acción/efectos de los fármacos , Animales , Butadienos/farmacología , Duramadre/fisiopatología , Estimulación Eléctrica , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hiperalgesia/complicaciones , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Interleucina-6/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Meninges/efectos de los fármacos , Meninges/patología , Trastornos Migrañosos/complicaciones , Trastornos Migrañosos/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7 , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/enzimología , Nitrilos/farmacología , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Canales de Sodio/metabolismo
20.
Results Probl Cell Differ ; 54: 41-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22009347

RESUMEN

The purpose of this chapter is to discuss the role of the fragile X mental retardation protein (FMRP) in the spinal sensory system and the potential for use of the mouse model of fragile X syndrome to better understand some aspects of the human syndrome as well as advance knowledge in other areas of investigation, such as pain amplification, an important aspect of clinical pain disorders. We describe how the Fmr1 knockout mouse can be used to better understand the role of Fmrp in axons using cultures of sensory neurons and using manipulations to these neurons in vivo. We also discuss the established evidence for a role of Fmrp in nociceptive sensitization and how this evidence relates to an emerging role of translation control as a key process in pain amplification. Finally, we explore opportunities centered on the Fmr1 KO mouse for gaining further insight into the role of translation control in pain amplification and how this model may be used to identify novel therapeutic targets. We conclude that the study of the spinal sensory system in the Fmr1 KO mouse presents several unique prospects for gaining better insight into the human disorder and other clinical issues, such as chronic pain disorders, that affect millions of people worldwide.


Asunto(s)
Dolor Crónico/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Células Receptoras Sensoriales/fisiología , Médula Espinal/fisiología , Animales , Axones/fisiología , Conducta Animal/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Humanos , Ratones , Ratones Noqueados , Plasticidad Neuronal/fisiología , Células Receptoras Sensoriales/citología , Médula Espinal/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA