Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Anesthesiology ; 139(4): 476-491, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37351557

RESUMEN

BACKGROUND: Chronic postsurgical pain is a poorly recognized outcome of surgery where patients experience pain long after healing from the surgical insult. Descending control of nociception, a phenomenon whereby application of a strong nociceptive stimulus to one part of the body of animals inhibits pain in remote body regions, offers one strategy to identify a propensity to develop chronic postsurgical pain-like behavior. Here, consomic rat panel was used to test the hypothesis that pain persistence is mechanistically linked to ineffective descending control of nociception. METHODS: Male and female Brown Norway, Dahl S, and eight consomic strains (SS-xBN) were used to determine the presence of chronic postsurgical pain-like behaviors by using paw-withdrawal threshold evaluation (von Frey method) in the area adjacent to a hind paw plantar incision. Descending control of nociception was assessed by measuring hind paw-withdrawal thresholds (Randall-Selitto method) after capsaicin (125 µg) injection into a forepaw. Consomic rats were developed by introgressing individual Brown Norway chromosomes on the Dahl S rat genetic background, as Dahl S rats lack preoperative descending control of nociception. RESULTS: Substitution of several chromosomes from the "pain-resistant" Brown Norway to the "pain-prone" Dahl S/Medical College of Wisconsin reduced mechanical nociceptive sensitivity and increased endogenous pain modulation capacity by differing degrees. Statistical modeling of these data revealed that descending control of nociception is a poor general predictor of the propensity to develop chronic postsurgical pain-like behavior (poor fit for model 1). However, a significant strain-by-descending control of nociception interaction was revealed (model 3, -2*log likelihood; 550.668, -2ll change; 18.093, P = 0.034) with SS-13BN and SS-15BN strains showing a negative descending control of nociception relationship with chronic postsurgical pain-like behavior. CONCLUSIONS: Descending control of nociception poorly predicted which rat strains developed chronic postsurgical pain-like behavior despite controlling for genetic, environmental, and sex differences. Two consomic strains that mimic clinical chronic postsurgical pain criteria and display a strong negative correlation with descending control of nociception were identified, offering novel candidates for future experiments exploring mechanisms that lead to chronic postsurgical pain.


Asunto(s)
Cromosomas , Nocicepción , Ratas , Animales , Femenino , Masculino , Ratas Endogámicas BN , Ratas Endogámicas Dahl , Dolor Postoperatorio/genética
2.
Environ Monit Assess ; 195(6): 704, 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37212972

RESUMEN

Sustainable use of groundwater while maintaining economic and social development is a major challenge, and the implementation of wellhead protection areas (WHPA) for public supply wells has been applied as an instrument to overcome it. This study analyzes the WHPA delineation methods: calculated fixed radius (CFR) and two solutions of the WhAEM software (USEPA, 2018), one analytical and one semi-analytical. We compare their results with WHPAs generated by a stochastic three-dimensional MODFLOW-MODPATH model in two scenarios: eight pumping wells operating simultaneously and a single well pumping, both at the same public drinking water supply wellfield located on a coastal plain in Jaguaruna County, south Brazil. For the specific hydrogeological settings, all methods produced satisfactory results when delineating a 50-day time-of-travel (TOT) WHPA for a single well. However, as TOT increases, uncertainties are introduced, and the precision of the results is reduced. Multiple well pumping simultaneously presented similar issues regarding uncertainties caused by three-dimensional flow complexities resulting from well interferences. Despite being the simplest method applied in terms of hydrogeological data needs, the CFR method demonstrated reliability in its results. Additionally, we present an analysis comparing the dimensions of the capture zone with the 10- and 20-year TOT WHPAs, indicating that managing the entire capture zone is the best way to protect groundwater against conservative contaminants. Finally, we compare WHPA generated by a stochastic and a deterministic model to understand how uncertainties can affect model results.


Asunto(s)
Monitoreo del Ambiente , Agua Subterránea , Reproducibilidad de los Resultados , Monitoreo del Ambiente/métodos , Abastecimiento de Agua , Pozos de Agua , Modelos Teóricos , Movimientos del Agua
3.
J Neurochem ; 157(6): 1821-1837, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32885411

RESUMEN

Hyperalgesic priming is characterized by enhanced nociceptor sensitization by pronociceptive mediators, prototypically PGE2 . Priming has gained interest as a mechanism underlying the transition to chronic pain. Which stimuli induce priming and what cellular mechanisms are employed remains incompletely understood. In adult male rats, we present the cytokine Oncostatin M (OSM), a member of the IL-6 family, as an inducer of priming by a novel mechanism. We used a high content microscopy based approach to quantify the activation of endogenous PKA-II and ERK of thousands sensory neurons in culture. Incubation with OSM increased and prolonged ERK activation by agents that increase cAMP production such as PGE2 , forskolin, and cAMP analogs. These changes were specific to IB4/CaMKIIα positive neurons, required protein translation, and increased cAMP-to-ERK signaling. In both, control and OSM-treated neurons, cAMP/ERK signaling involved RapGEF2 and PKA but not Epac. Similar enhancement of cAMP-to-ERK signaling could be induced by GDNF, which acts mostly on IB4/CaMKIIα-positive neurons, but not by NGF, which acts mostly on IB4/CaMKIIα-negative neurons. In vitro, OSM pretreatment rendered baseline TTX-R currents ERK-dependent and switched forskolin-increased currents from partial to full ERK-dependence in small/medium sized neurons. In summary, priming induced by OSM uses a novel mechanism to enhance and prolong coupling of cAMP/PKA to ERK1/2 signaling without changing the overall pathway structure.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hiperalgesia/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oncostatina M/toxicidad , Animales , Antineoplásicos/toxicidad , Humanos , Hiperalgesia/inducido químicamente , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
4.
J Neurosci ; 38(2): 308-321, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29175954

RESUMEN

We studied, in male Sprague Dawley rats, the role of the cognate hyaluronan receptor, CD44 signaling in the antihyperalgesia induced by high molecular weight hyaluronan (HMWH). Low molecular weight hyaluronan (LMWH) acts at both peptidergic and nonpeptidergic nociceptors to induce mechanical hyperalgesia that is prevented by intrathecal oligodeoxynucleotide antisense to CD44 mRNA, which also prevents hyperalgesia induced by a CD44 receptor agonist, A6. Ongoing LMWH and A6 hyperalgesia are reversed by HMWH. HMWH also reverses the hyperalgesia induced by diverse pronociceptive mediators, prostaglandin E2, epinephrine, TNFα, and interleukin-6, and the neuropathic pain induced by the cancer chemotherapy paclitaxel. Although CD44 antisense has no effect on the hyperalgesia induced by inflammatory mediators or paclitaxel, it eliminates the antihyperalgesic effect of HMWH. HMWH also reverses the hyperalgesia induced by activation of intracellular second messengers, PKA and PKCε, indicating that HMWH-induced antihyperalgesia, although dependent on CD44, is mediated by an intracellular signaling pathway rather than as a competitive receptor antagonist. Sensitization of cultured small-diameter DRG neurons by prostaglandin E2 is also prevented and reversed by HMWH. These results demonstrate the central role of CD44 signaling in HMWH-induced antihyperalgesia, and establish it as a therapeutic target against inflammatory and neuropathic pain.SIGNIFICANCE STATEMENT We demonstrate that hyaluronan (HA) with different molecular weights produces opposing nociceptive effects. While low molecular weight HA increases sensitivity to mechanical stimulation, high molecular weight HA reduces sensitization, attenuating inflammatory and neuropathic hyperalgesia. Both pronociceptive and antinociceptive effects of HA are mediated by activation of signaling pathways downstream CD44, the cognate HA receptor, in nociceptors. These results contribute to our understanding of the role of the extracellular matrix in pain, and indicate CD44 as a potential therapeutic target to alleviate inflammatory and neuropathic pain.


Asunto(s)
Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Hiperalgesia/metabolismo , Nociceptores/metabolismo , Animales , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Masculino , Peso Molecular , Ratas , Ratas Sprague-Dawley , Transducción de Señal
5.
J Neurosci ; 38(9): 2226-2245, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29431655

RESUMEN

Systemic fentanyl induces hyperalgesic priming, long-lasting neuroplasticity in nociceptor function characterized by prolongation of inflammatory mediator hyperalgesia. To evaluate priming at both nociceptor terminals, we studied, in male Sprague Dawley rats, the effect of local administration of agents that reverse type I (protein translation) or type II [combination of Src and mitogen-activated protein kinase (MAPK)] priming. At the central terminal, priming induced by systemic, intradermal, or intrathecal fentanyl was reversed by the combination of Src and MAPK inhibitors, but at the peripheral terminal, it was reversed by the protein translation inhibitor. Mu-opioid receptor (MOR) antisense prevented fentanyl hyperalgesia and priming. To determine whether type I and II priming occur in the same population of neurons, we used isolectin B4-saporin or [Sar9, Met(O2)11]-substance P-saporin to deplete nonpeptidergic or peptidergic nociceptors, respectively. Following intrathecal fentanyl, central terminal priming was prevented by both saporins, whereas that in peripheral terminal was not attenuated even by their combination. However, after intradermal fentanyl, priming in the peripheral terminal requires both peptidergic and nonpeptidergic nociceptors, whereas that in the central terminal is dependent only on peptidergic nociceptors. Pretreatment with dantrolene at either terminal prevented fentanyl-induced priming in both terminals, suggesting communication between central and peripheral terminals mediated by intracellular Ca2+ signaling. In vitro application of fentanyl increased cytoplasmic Ca2+ concentration in dorsal root ganglion neurons, which was prevented by pretreatment with dantrolene and naloxone. Therefore, acting at MOR in the nociceptor, fentanyl induces hyperalgesia and priming rapidly at both the central (type II) and peripheral (type I) terminal and this is mediated by Ca2+ signaling.SIGNIFICANCE STATEMENT Fentanyl, acting at the µ-opioid receptor (MOR), induces hyperalgesia and hyperalgesic priming at both the central and peripheral terminal of nociceptors and this is mediated by endoplasmic reticulum Ca2+ signaling. Priming in the central terminal is type II, whereas that in the peripheral terminal is type I. Our findings may provide useful information for the design of drugs with improved therapeutic profiles, selectively disrupting individual MOR signaling pathways, to maintain an adequate long-lasting control of pain.


Asunto(s)
Analgésicos Opioides/farmacología , Fentanilo/farmacología , Hiperalgesia/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Masculino , Nociceptores/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/fisiología
6.
J Neurosci ; 38(14): 3394-3413, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29483280

RESUMEN

Nerve growth factor (NGF) exerts multiple functions on target neurons throughout development. The recent discovery of a point mutation leading to a change from arginine to tryptophan at residue 100 in the mature NGFß sequence (NGFR100W) in patients with hereditary sensory and autonomic neuropathy type V (HSAN V) made it possible to distinguish the signaling mechanisms that lead to two functionally different outcomes of NGF: trophic versus nociceptive. We performed extensive biochemical, cellular, and live-imaging experiments to examine the binding and signaling properties of NGFR100W Our results show that, similar to the wild-type NGF (wtNGF), the naturally occurring NGFR100W mutant was capable of binding to and activating the TrkA receptor and its downstream signaling pathways to support neuronal survival and differentiation. However, NGFR100W failed to bind and stimulate the 75 kDa neurotrophic factor receptor (p75NTR)-mediated signaling cascades (i.e., the RhoA-Cofilin pathway). Intraplantar injection of NGFR100W into adult rats induced neither TrkA-mediated thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia based on agonism for TrkA signaling. Together, our studies provide evidence that NGFR100W retains trophic support capability through TrkA and one aspect of its nociceptive signaling, but fails to engage p75NTR signaling pathways. Our findings suggest that wtNGF acts via TrkA to regulate the delayed priming of nociceptive responses. The integration of both TrkA and p75NTR signaling thus appears to regulate neuroplastic effects of NGF in peripheral nociception.SIGNIFICANCE STATEMENT In the present study, we characterized the naturally occurring nerve growth factor NGFR100W mutant that is associated with hereditary sensory and autonomic neuropathy type V. We have demonstrated for the first time that NGFR100W retains trophic support capability through TrkA, but fails to engage p75NTR signaling pathways. Furthermore, after intraplantar injection into adult rats, NGFR100W induced neither thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia. We have also provided evidence that the integration of both TrkA- and p75NTR-mediated signaling appears to regulate neuroplastic effects of NGF in peripheral nociception. Our study with NGFR100W suggests that it is possible to uncouple trophic effect from nociceptive function, both induced by wild-type NGF.


Asunto(s)
Neuropatías Hereditarias Sensoriales y Autónomas/genética , Mutación Missense , Factor de Crecimiento Nervioso/genética , Nocicepción , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células 3T3 , Animales , Células Cultivadas , Células HEK293 , Neuropatías Hereditarias Sensoriales y Autónomas/metabolismo , Neuropatías Hereditarias Sensoriales y Autónomas/fisiopatología , Humanos , Masculino , Ratones , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso , Células PC12 , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento , Transducción de Señal
7.
J Neurosci ; 37(8): 2032-2044, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28115480

RESUMEN

Hyperalgesic priming, a model of pain chronification in the rat, is mediated by ryanodine receptor-dependent calcium release. Although ryanodine induces priming in both sexes, females are 5 orders of magnitude more sensitive, by an estrogen receptor α (EsRα)-dependent mechanism. An inositol 1,4,5-triphosphate (IP3) receptor inhibitor prevented the induction of priming by ryanodine. For IP3 induced priming, females were also more sensitive. IP3-induced priming was prevented by pretreatment with inhibitors of the sarcoendoplasmic reticulum calcium ATPase and ryanodine receptor. Antisense to EsRα prevented the induction of priming by low-dose IP3 in females. The induction of priming by an EsRα agonist was ryanodine receptor-dependent and prevented by the IP3 antagonist. Thus, an EsRα-dependent bidirectional interaction between endoplasmic reticulum IP3 and ryanodine receptor-mediated calcium signaling is present in the induction of hyperalgesic priming, in females. In cultured male DRG neurons, IP3 (100 µm) potentiated depolarization-induced transients produced by extracellular application of high-potassium solution (20 mm, K20), in nociceptors incubated with ß-estradiol. This potentiation of depolarization-induced calcium transients was blocked by the IP3 antagonist, and not observed in the absence of IP3 IP3 potentiation was also blocked by ryanodine receptor antagonist. The application of ryanodine (2 nm), instead of IP3, also potentiated K20-induced calcium transients in the presence of ß-estradiol, in an IP3 receptor-dependent manner. Our results point to an EsRα-dependent, reciprocal interaction between IP3 and ryanodine receptors that contributes to sex differences in hyperalgesic priming.SIGNIFICANCE STATEMENT The present study demonstrates a mechanism that plays a role in the marked sexual dimorphism observed in a model of the transition to chronic pain, hyperalgesic priming. This mechanism involves a reciprocal interaction between the endoplasmic reticulum receptors, IP3 and ryanodine, in the induction of priming, regulated by estrogen receptor α in the nociceptor of female rats. The presence of this signaling pathway modulating the susceptibility of nociceptors to develop plasticity may contribute to our understanding of sex differences observed clinically in chronic pain syndromes.


Asunto(s)
Hiperalgesia/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Umbral del Dolor/fisiología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Caracteres Sexuales , Animales , Células Cultivadas , Dinoprostona/efectos adversos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Ganglios Espinales/citología , Hiperalgesia/inducido químicamente , Inositol 1,4,5-Trifosfato/farmacología , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Compuestos Macrocíclicos/farmacología , Masculino , Oligodesoxirribonucleótidos Antisentido/farmacología , Oxazoles/farmacología , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Rianodina/efectos adversos , Células Receptoras Sensoriales/efectos de los fármacos , Tapsigargina/farmacología
8.
Proc Natl Acad Sci U S A ; 111(51): 18363-8, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25489099

RESUMEN

The present study evaluated the role of N-methyl-D-aspartate receptors (NMDARs) expressed in the dorsal root ganglia (DRG) in the inflammatory sensitization of peripheral nociceptor terminals to mechanical stimulation. Injection of NMDA into the fifth lumbar (L5)-DRG induced hyperalgesia in the rat hind paw with a profile similar to that of intraplantar injection of prostaglandin E2 (PGE2), which was significantly attenuated by injection of the NMDAR antagonist D(-)-2-amino-5-phosphonopentanoic acid (D-AP-5) in the L5-DRG. Moreover, blockade of DRG AMPA receptors by the antagonist 6,7-dinitroquinoxaline-2,3-dione had no effect in the PGE2-induced hyperalgesia in the paw, showing specific involvement of NMDARs in this modulatory effect and suggesting that activation of NMDAR in the DRG plays an important role in the peripheral inflammatory hyperalgesia. In following experiments we observed attenuation of PGE2-induced hyperalgesia in the paw by the knockdown of NMDAR subunits NR1, NR2B, NR2D, and NR3A with antisense-oligodeoxynucleotide treatment in the DRG. Also, in vitro experiments showed that the NMDA-induced sensitization of cultured DRG neurons depends on satellite cell activation and on those same NMDAR subunits, suggesting their importance for the PGE2-induced hyperalgesia. In addition, fluorescent calcium imaging experiments in cultures of DRG cells showed induction of calcium transients by glutamate or NMDA only in satellite cells, but not in neurons. Together, the present results suggest that the mechanical inflammatory nociceptor sensitization is dependent on glutamate release at the DRG and subsequent NMDAR activation in satellite glial cells, supporting the idea that the peripheral hyperalgesia is an event modulated by a glutamatergic system in the DRG.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Nociceptores/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Células Satélites Perineuronales/efectos de los fármacos , 2-Amino-5-fosfonovalerato/farmacología , Animales , Dinoprostona/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Quinoxalinas/farmacología , Ratas , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Células Satélites Perineuronales/metabolismo
9.
J Neurosci ; 35(36): 12502-17, 2015 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-26354917

RESUMEN

The primary afferent nociceptor was used as a model system to study mechanisms of pain induced by chronic opioid administration. Repeated intradermal injection of the selective mu-opioid receptor (MOR) agonist DAMGO induced mechanical hyperalgesia and marked prolongation of prostaglandin E2 (PGE2) hyperalgesia, a key feature of hyperalgesic priming. However, in contrast to prior studies of priming induced by receptor-mediated (i.e., TNFα, NGF, or IL-6 receptor) or direct activation of protein kinase Cε (PKCε), the pronociceptive effects of PGE2 in DAMGO-treated rats demonstrated the following: (1) rapid induction (4 h compared with 3 d); (2) protein kinase A (PKA), rather than PKCε, dependence; (3) prolongation of hyperalgesia induced by an activator of PKA, 8-bromo cAMP; (4) failure to be reversed by a protein translation inhibitor; (5) priming in females as well as in males; and (6) lack of dependence on the isolectin B4-positive nociceptor. These studies demonstrate a novel form of hyperalgesic priming induced by repeated administration of an agonist at the Gi-protein-coupled MOR to the peripheral terminal of the nociceptor. Significance statement: The current study demonstrates the molecular mechanisms involved in the sensitization of nociceptors produced by repeated activation of mu-opioid receptors and contributes to our understanding of the painful condition observed in patients submitted to chronic use of opioids.


Asunto(s)
Dolor Crónico/metabolismo , Hiperalgesia/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Femenino , Glicoproteínas/metabolismo , Lectinas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Versicanos
10.
J Neurosci ; 35(15): 6107-16, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878283

RESUMEN

Hyperalgesic priming, a form of neuroplasticity in nociceptors, is a model of the transition from acute to chronic pain in the rat, which involves signaling from the site of an acute tissue insult in the vicinity of the peripheral terminal of a nociceptor to its cell body that, in turn, induces a signal that travels back to the terminal to mediate a marked prolongation of prostaglandin E2-induced hyperalgesia. In the present experiments, we studied the underlying mechanisms in the cell body and compared them to the mechanisms in the nerve terminal. Injection of a cell-permeant cAMP analog, 8-bromo cAMP, into the dorsal root ganglion induced mechanical hyperalgesia and priming with an onset more rapid than when induced at the peripheral terminal. Priming induced by intraganglion 8-bromo cAMP was prevented by an oligodeoxynucleotide antisense to mRNA for a transcription factor, cAMP response element-binding protein (CREB), and by an inhibitor of importin, which is required for activated CREB to get into the nucleus. While peripheral administration of 8-bromo cAMP also produced hyperalgesia, it did not produce priming. Conversely, interventions administered in the vicinity of the peripheral terminal of the nociceptor that induces priming-PKCε activator, NGF, and TNF-α-when injected into the ganglion produce hyperalgesia but not priming. The protein translation inhibitor cordycepin, injected at the peripheral terminal but not into the ganglion, reverses priming induced at either the ganglion or peripheral terminal of the nociceptor. These data implicate different mechanisms in the soma and terminal in the transition to chronic pain.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Hiperalgesia/etiología , Hiperalgesia/patología , Nociceptores/citología , Umbral del Dolor/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/toxicidad , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Quimiocina CCL2/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Dactinomicina/metabolismo , Dinoprostona/toxicidad , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Ganglios Espinales/citología , Hiperalgesia/prevención & control , Masculino , Nociceptores/efectos de los fármacos , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Umbral del Dolor/efectos de los fármacos , Estimulación Física/efectos adversos , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley
11.
J Neurosci ; 35(2): 495-507, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589745

RESUMEN

Acute insults produce hyperalgesic priming, a neuroplastic change in nociceptors that markedly prolongs inflammatory mediator-induced hyperalgesia. After an acute initiating insult, there is a 72 h delay to the onset of priming, for which the underlying mechanism is unknown. We hypothesized that the delay is due to the time required for a signal to travel from the peripheral terminal to the cell body followed by a return signal to the peripheral terminal. We report that when an inducer of hyperalgesic priming (monocyte chemotactic protein 1) is administered at the spinal cord of Sprague Dawley rats, priming is detected at the peripheral terminal with a delay significantly shorter than when applied peripherally. Spinally induced priming is detected not only when prostaglandin E2 (PGE2) is presented to the peripheral nociceptor terminals, but also when it is presented intrathecally to the central terminals in the spinal cord. Furthermore, when an inducer of priming is administered in the paw, priming can be detected in spinal cord (as prolonged hyperalgesia induced by intrathecal PGE2), but only when the mechanical stimulus is presented to the paw on the side where the priming inducer was administered. Both spinally and peripherally induced priming is prevented by intrathecal oligodeoxynucleotide antisense to the nuclear transcription factor CREB mRNA. Finally, the inhibitor of protein translation reversed hyperalgesic priming only when injected at the site where PGE2 was administered, suggesting that the signal transmitted from the cell body to the peripheral terminal is not a newly translated protein, but possibly a newly expressed mRNA.


Asunto(s)
Dolor Agudo/fisiopatología , Axones/fisiología , Dolor Crónico/fisiopatología , Nociceptores/fisiología , Tiempo de Reacción , Dolor Agudo/metabolismo , Animales , Axones/metabolismo , Quimiocina CCL2/farmacología , Dolor Crónico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dinoprostona/farmacología , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología
12.
Proc Natl Acad Sci U S A ; 110(9): 3603-8, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23401543

RESUMEN

It is well established that dorsal root ganglion (DRG) cells synthesize prostaglandin. However, the role that prostaglandin plays in the inflammatory hyperalgesia of peripheral tissue has not been established. Recently, we have successfully established a technique to inject drugs (3 µL) directly into the L5-DRG of rats, allowing in vivo identification of the role that DRG cell-derived COX-1 and COX-2 play in the development of inflammatory hyperalgesia of peripheral tissue. IL-1ß (0.5 pg) or carrageenan (100 ng) was administered in the L5-peripheral field of rat hindpaw and mechanical hyperalgesia was evaluated after 3 h. Administration of a nonselective COX inhibitor (indomethacin), selective COX-1 (valeryl salicylate), or selective COX-2 (SC-236) inhibitors into the L5-DRG prevented the hyperalgesia induced by IL-1ß. Similarly, oligodeoxynucleotide-antisense against COX-1 or COX-2, but not oligodeoxynucleotide-mismatch, decreased their respective expressions in the L5-DRG and prevented the hyperalgesia induced by IL-1ß in the hindpaw. Immunofluorescence analysis demonstrated that the amount of COX-1 and COX-2, constitutively expressed in TRPV-1(+) cells of the DRG, significantly increased after carrageenan or IL-1ß administration. In addition, indomethacin administered into the L5-DRG prevented the increase of PKCε expression in DRG membrane cells induced by carrageenan. Finally, the administration of EP1/EP2 (7.5 ng) or EP4 (10 µg) receptor antagonists into L5-DRG prevented the hyperalgesia induced by IL-1ß in the hindpaw. In conclusion, the results of this study suggest that the inflammatory hyperalgesia in peripheral tissue depends on activation of COX-1 and COX-2 in C-fibers, which contribute to the induction and maintenance of sensitization of primary sensory neurons.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Ganglios Espinales/enzimología , Hiperalgesia/enzimología , Hiperalgesia/patología , Inflamación/enzimología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Animales , Carragenina/farmacología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/farmacología , Activación Enzimática/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Técnicas de Silenciamiento del Gen , Hiperalgesia/complicaciones , Indometacina/administración & dosificación , Indometacina/farmacología , Inflamación/complicaciones , Interleucina-1beta/farmacología , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Masculino , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/metabolismo
13.
Mol Pain ; 11: 65, 2015 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-26497944

RESUMEN

BACKGROUND: We evaluated the role of a mechanically-gated ion channel, Piezo2, in mechanical stimulation-induced enhancement of hyperalgesia produced by the pronociceptive vasoactive mediator endothelin-1, an innocuous mechanical stimulus-induced enhancement of hyperalgesia that is vascular endothelial cell dependent. We also evaluated its role in a preclinical model of a vascular endothelial cell dependent painful peripheral neuropathy. RESULTS: The local administration of oligodeoxynucleotides antisense to Piezo2 mRNA, at the site of nociceptive testing in the rat's hind paw, but not intrathecally at the central terminal of the nociceptor, prevented innocuous stimulus-induced enhancement of hyperalgesia produced by endothelin-1 (100 ng). The mechanical hyperalgesia induced by oxaliplatin (2 mg/kg. i.v.), which was inhibited by impairing endothelial cell function, was similarly attenuated by local injection of the Piezo2 antisense. Polymerase chain reaction analysis demonstrated for the first time the presence of Piezo2 mRNA in endothelial cells. CONCLUSIONS: These results support the hypothesis that Piezo2 is a mechano-transducer in the endothelial cell where it contributes to stimulus-dependent hyperalgesia, and a model of chemotherapy-induced painful peripheral neuropathy.


Asunto(s)
Células Endoteliales/patología , Canales Iónicos/metabolismo , Dolor/fisiopatología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Animales , Antineoplásicos/farmacología , Células Endoteliales/efectos de los fármacos , Endotelina-1/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Canales Iónicos/genética , Masculino , Modelos Animales , Oligonucleótidos Antisentido/farmacología , Compuestos Organoplatinos/farmacología , Oxaliplatino , Dolor/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Ratas , Ratas Sprague-Dawley , Tacto
14.
J Neurosci ; 33(27): 11002-11, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23825405

RESUMEN

We have previously shown that activation of protein kinase Cε (PKCε) in male rats induces a chronic, long-lasting change in nociceptors such that a subsequent exposure to proinflammatory mediators produces markedly prolonged mechanical hyperalgesia. This neuroplastic change, hyperalgesic priming, is dependent on activation of cytoplasmic polyadenylation element-binding protein (CPEB), downstream of PKCε, and consequent translation of mRNAs in the peripheral terminal of the nociceptor. Since α calmodulin-dependent protein kinase II (αCaMKII), a molecule implicated in neuroplasticity, is a target of CPEB and can also affect CPEB function, we investigated its role in the transition from acute to chronic pain. Priming induced by direct activation of PKCε can be prevented by inhibition of αCaMKII. In addition, direct activation of αCaMKII induces priming, which was not prevented by pretreatment with PKCε antisense, suggesting that αCaMKII is downstream of PKCε in the induction of priming. Activation of ryanodine receptors (RyRs), which can lead to activation of αCaMKII, also induced priming, in a calcium- and αCaMKII-dependent manner. Similarly, inhibition of the RyR and a calcium buffer prevented induction of priming by PKCε. Unlike activation of PKCε, ryanodine and αCaMKII induced priming in female as well as male rats. Our results demonstrate a contribution of αCaMKII to induction of hyperalgesic priming, a phenomenon implicated in the transition from acute to chronic pain.


Asunto(s)
Dolor Agudo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Dolor Crónico/metabolismo , Hiperalgesia/metabolismo , Nociceptores/fisiología , Dimensión del Dolor/métodos , Dolor Agudo/patología , Animales , Dolor Crónico/patología , Femenino , Hiperalgesia/patología , Masculino , Ratas , Ratas Sprague-Dawley
15.
Sci Total Environ ; 912: 168958, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38029979

RESUMEN

Groundwater storage changes in the Amazon River Basin (ARB) play an important role in the hydrological behavior of the region, with significant influence on climate variability and rainforest ecosystems. The GRACE and GRACE-FO satellite missions provide gravity anomalies from which it is possible to monitor changes in terrestrial water storage, albeit at low spatial resolution. This study downscaled GRACE and GRACE-FO data from machine learning models from 1° (110 km approx) to 0.25° (27.5 km approx). It estimated the spatiotemporal variability of terrestrial and groundwater storage anomalies between 2002 and 2021 for the Amazon River Basin. In parallel, the Random Forest and AdaBoost algorithms were compared and analyzed. The results reflected a good fit of the models with a very low error and a slight superiority in the predictions obtained by AdaBoost. On the predictions at 0.25°, spatial patterns associated with the strong influence on storage changes of some rivers and snow-capped mountains were identified, as well as an increase in the accuracy of the scaled data of the original ones. Positive long-term behavior was also obtained in terrestrial and groundwater storage of 14.26 ± 1.18 km3/yr and + 22.24 ± 1.18 km3/yr, respectively. Validation of the time series of groundwater anomalies to water levels in the monitoring wells obtained maximum correlation coefficients of 0.85 with confidence levels of 0.01. These results are promising for satellite information in water management, especially in regional monitoring of unconfined aquifers. The obtained data is stored in a dedicated repository (Satizábal-Alarcón et al., 2023).

16.
Eur J Neurosci ; 37(10): 1705-13, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23379641

RESUMEN

We have previously shown, in the rat, that neuropathic and inflammatory events produce a neuroplastic change in nociceptor function whereby a subsequent exposure to a proinflammatory mediator (e.g. prostaglandin E2 ; PGE2 ) produces markedly prolonged mechanical hyperalgesia. While the initial approximately 30 min of this prolonged PGE2 hyperalgesia remains PKA-dependent, it subsequently switches to become dependent on protein kinase C epsilon (PKCε). In this study we tested the hypothesis that the delayed onset, PKCε-mediated, component of PGE2 hyperalgesia is generated by the active release of a nucleotide from the peripheral terminal of the primed nociceptor and this nucleotide is then metabolized to produce adenosine, which acts on a Gi-coupled A1 adenosine receptor on the nociceptor to generate PKCε-dependent hyperalgesia. We report that inhibitors of ATP-binding cassette transporters, of ecto-5'-phosphodiesterase and ecto-5'nucleotidase (enzymes involved in the metabolism of cyclic nucleotides to adenosine) and of A1 adenosine receptors each eliminated the late, but not the early, phase of PGE2 -induced hyperalgesia in primed animals. A second model of chronic pain induced by transient attenuation of G-protein-coupled receptor kinase 2, in which the prolongation of PGE2 hyperalgesia is not PKCε-dependent, was not attenuated by inhibitors of any of these mechanisms. Based on these results we propose a contribution of an autocrine mechanism, in the peripheral terminal of the nociceptor, in the hyperalgesic priming model of chronic pain.


Asunto(s)
Comunicación Autocrina , Dolor Crónico/metabolismo , Nociceptores/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Dinoprostona/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Hiperalgesia/metabolismo , Masculino , Nociceptores/efectos de los fármacos , Proteína Quinasa C-epsilon/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A1/metabolismo
17.
J Clin Invest ; 133(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36856117

RESUMEN

Only three classes of pain medications have made it into clinical use in the past 60 years despite intensive efforts and the need for nonaddictive pain treatments. One reason for the failure involves the use of animal models that lack mechanistic similarity to human pain conditions, with endpoint measurements that may not reflect the human pain experience. In this issue of the JCI, Ding, Fischer, and co-authors developed the foramen lacerum impingement of trigeminal nerve root (FLIT) model of human trigeminal neuralgia that has improved face, construct, and predictive validities over those of current models. They used the FLIT model to investigate the role that abnormal, hypersynchronous cortical activity contributed to a neuropathic pain state. Unrestrained, synchronous glutamatergic activity in the primary somatosensory cortex upper lip and jaw (S1ULp-S1J) region of the somatosensory cortex drove pain phenotypes. The model establishes a powerful tool to continue investigating the interaction between the peripheral and central nervous systems that leads to chronic pain.


Asunto(s)
Dolor Crónico , Neuralgia , Animales , Humanos , Dolor Crónico/terapia , Sistema Nervioso Central , Neuralgia/terapia , Modelos Animales , Manejo del Dolor
18.
J Neurosci ; 31(31): 11404-10, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21813700

RESUMEN

While oxidative stress has been implicated in small-fiber painful peripheral neuropathies, antioxidants are only partially effective to treat patients. We have tested the hypothesis that Drp1 (dynamin-related protein 1), a GTPase that catalyzes the process of mitochondrial fission, which is a mechanism central for the effect and production of reactive oxygen species (ROS), plays a central role in these neuropathic pain syndromes. Intrathecal administration of oligodeoxynucleotide antisense against Drp1 produced a decrease in its expression in peripheral nerve and markedly attenuated neuropathic mechanical hyperalgesia caused by HIV/AIDS antiretroviral [ddC (2',3'-dideoxycytidine)] and anticancer (oxaliplatin) chemotherapy in male Sprague Dawley rats. To confirm the role of Drp1 in these models of neuropathic pain, as well as to demonstrate its contribution at the site of sensory transduction, we injected a highly selective Drp1 inhibitor, mdivi-1, at the site of nociceptive testing on the dorsum of the rat's hindpaw. mdivi-1 attenuated both forms of neuropathic pain. To evaluate the role of Drp1 in hyperalgesia induced by ROS, we demonstrated that intradermal hydrogen peroxide produced dose-dependent hyperalgesia that was inhibited by mdivi-1. Finally, mechanical hyperalgesia induced by diverse pronociceptive mediators involved in inflammatory and neuropathic pain-tumor necrosis factor α, glial-derived neurotrophic factor, and nitric oxide-was also inhibited by mdivi-1. These studies provide support for a substantial role of mitochondrial fission in preclinical models of inflammatory and neuropathic pain.


Asunto(s)
Dinaminas/metabolismo , Neuralgia/metabolismo , Análisis de Varianza , Animales , Fármacos Anti-VIH/toxicidad , Antineoplásicos/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Dinaminas/genética , Epinefrina/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Peróxido de Hidrógeno , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Masculino , Factor de Crecimiento Nervioso/uso terapéutico , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Donantes de Óxido Nítrico/toxicidad , Nitrocompuestos/toxicidad , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Dimensión del Dolor/métodos , Quinazolinonas/uso terapéutico , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/uso terapéutico , Zalcitabina/toxicidad
19.
Ann Neurol ; 70(1): 101-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21786301

RESUMEN

OBJECTIVE: While inflammatory pain is well described in skeletal muscle, neuropathic muscle pain remains to be clarified. We used 3 well-established rodent models of peripheral neuropathy to evaluate for muscle pain. METHODS: In rats exposed to either of 2 neurotoxic cancer chemotherapies, paclitaxel or oxaliplatin, or to alcohol consumption, we assessed the evolution of mechanical hyperalgesia in skeletal muscle and skin, in the same animal. To explore the involvement of protein kinase C epsilon (PKCε), a second messenger implicated in some forms of neuropathic pain, antisense oligodeoxynucleotides (AS-ODNs) or mismatch ODNs (MM-ODNs) for PKCε were administered intrathecally. RESULTS: Rats submitted to models of chemotherapy-induced and alcohol-induced neuropathy developed persistent muscle hyperalgesia, which evolved in parallel in muscle and skin. The administration of PKCε AS, which has been shown to mediate cutaneous hyperalgesia in paclitaxel and ethanol models of neuropathic pain, also inhibited muscle hyperalgesia induced by these agents. Stopping AS-ODN was associated with the reappearance of hyperalgesia at both sites. The AS-ODN to PKCε treatment was devoid of effect in both muscle and skin in the oxaliplatin neuropathy model. INTERPRETATION: Our results support the suggestion that neuropathic muscle pain may be a greater clinical problem than generally appreciated.


Asunto(s)
Neuropatía Alcohólica/patología , Antineoplásicos/toxicidad , Modelos Animales de Enfermedad , Músculo Esquelético/patología , Enfermedades del Sistema Nervioso Periférico/patología , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/patología , Neuropatía Alcohólica/etiología , Animales , Masculino , Músculo Esquelético/efectos de los fármacos , Dolor/inducido químicamente , Dolor/etiología , Dolor/patología , Dimensión del Dolor/métodos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Ratas , Ratas Sprague-Dawley
20.
Eur Spine J ; 21(3): 537-45, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21947908

RESUMEN

INTRODUCTION: The symptoms of lumbar disc herniation, such as low back pain and sciatica, have been associated with local release of cytokines following the inflammatory process induced by the contact of the nucleus pulposus (NP) with the spinal nerve. MATERIAL AND METHODS: Using an animal experimental model of intervertebral disc herniation and behavioral tests to evaluate mechanical (electronic von Frey test) and thermal (Hargreaves Plantar test) hyperalgesia in the hind paw of rats submitted to the surgical model, this study aimed to detect in normal intervertebral disc the cytokines known to be involved in the mechanisms of inflammatory hyperalgesia, to observe if previous exposure of the intervertebral disc tissue to specific antibodies could affect the pain behavior (mechanical and thermal hyperalgesia) induced by the NP, and to observe the influence of the time of contact of the NP with the fifth lumbar dorsal root ganglion (L5-DRG) in the mechanical and thermal hyperalgesia. RESULTS: The cytokines present at highest concentrations in the rat NP were TNF-α, IL-1ß and CINC-1. Rats submitted to the disc herniation experimental model, in which a NP from the sacrococcygeal region is deposited over the right L5-DRG, showed increased mechanical and thermal hyperalgesia that lasted at least 7 weeks. When the autologous NP was treated with antibodies against the three cytokines found at highest concentrations in the NP (TNF-α, IL-1ß and CINC-1), there was decrease in both mechanical and thermal hyperalgesia in different time points, suggesting that each cytokine may be important for the hyperalgesia in different steps of the inflammatory process. The surgical remotion of the NP from herniated rats 1 week after the implantation reduced the hyperalgesia to the level similar to the control group. This reduction in the hyperalgesia was also observed in the group that had the NP removed 3 weeks after the implantation, although the intensity of the hyperalgesia did not decreased totally. The removal of the NP after 5 weeks did not changed the hyperalgesia observed in the hind paw, which suggests that the longer the contact of the NP with the DRG, the greater is the possibility of development of chronic pain. CONCLUSION: Together our results indicate that specific cytokines released during the inflammatory process induced by the herniated intervertebral disc play fundamental role in the development of the two modalities of hyperalgesia (mechanical and thermal) and that the maintenance of this inflammation may be the most important point for the chronification of the pain.


Asunto(s)
Citocinas/antagonistas & inhibidores , Hiperalgesia/inmunología , Hiperalgesia/patología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , Animales , Anticuerpos/fisiología , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/fisiología , Citocinas/fisiología , Hiperalgesia/prevención & control , Interleucina-10/antagonistas & inhibidores , Interleucina-10/fisiología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/fisiología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/fisiología , Disco Intervertebral/patología , Masculino , Ratas , Ratas Wistar , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA