Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256047

RESUMEN

We recently demonstrated that transient attenuation of Toll-like receptor 4 (TLR4) in dorsal root ganglion (DRG) neurons, can both prevent and reverse pain associated with chemotherapy-induced peripheral neuropathy (CIPN), a severe side effect of cancer chemotherapy, for which treatment options are limited. Given the reduced efficacy of opioid analgesics to treat neuropathic, compared to inflammatory, pain, the crosstalk between nociceptor TLR4 and mu-opioid receptors (MOR), and that MOR and TLR4 agonists induce hyperalgesic priming (priming), which also occurs in CIPN, we determined, using male rats, whether: i ) antisense knockdown of nociceptor MOR attenuates CIPN, ii ) and attenuates the priming associated with CIPN, and iii ) CIPN also produces opioid-induced hyperalgesia (OIH). We found that intrathecal MOR antisense prevents and reverses hyperalgesia induced by oxaliplatin and paclitaxel, two common clinical chemotherapy agents. Oxaliplatin induced-priming was also markedly attenuated by MOR antisense. Additionally, intradermal morphine, at a dose that does not affect nociceptive threshold in controls, exacerbates mechanical hyperalgesia (OIH) in rats with CIPN, suggesting the presence of OIH. This OIH associated with CIPN is inhibited by interventions that reverse Type II priming (the combination of an inhibitor of Src and mitogen-activated protein kinase (MAPK)), a MOR antagonist, as well as a TLR4 antagonist. Our findings support a role of nociceptor MOR in oxaliplatin-induced pain and priming. We propose that priming and OIH are central to the symptom burden in CIPN, contributing to its chronicity and the limited efficacy of opioid analgesics to treat neuropathic pain.Significance Statement It remains unknown why opioid analgesics are less effective against neuropathic pain, including that induced by cancer chemotherapy. Here we demonstrate a crucial role of nociceptor mu-opioid receptors (MOR) in chemotherapy-induced peripheral neuropathy (CIPN) pain and hyperalgesic priming (priming), the latter a mechanism involved in the transition from acute to chronic pain. In addition to neuropathic pain and priming, opioid-induced hyperalgesia (OIH) also develops in chemotherapy-treated rats, which is reversed by inhibitors of second messengers that also reverse Type II priming, as well as by MOR and TLR4 antagonists. Taken together our results support the suggestion that priming and OIH contribute to CIPN and its limited responsiveness to opioid analgesics.

2.
Res Sq ; 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38464172

RESUMEN

The primary cilium, a 1-3 µm long hair-like structure protruding from the surface of almost all cells in the vertebrate body, is critical for neuronal development and also functions in the adult. As the migratory neural crest settles into dorsal root ganglia (DRG) sensory neurons elaborate a single primary cilium at their soma that is maintained into adult stages. While it is not known if primary cilia are expressed in nociceptors, or their potential function in the mature DRG neuron, recent studies have shown a role for Hedgehog, whose signaling demonstrates a dependence on primary cilia, in nociceptor sensitization. Here we report the expression of primary cilia in rat and mouse nociceptors, where they modulate mechanical nociceptive threshold, and contribute to inflammatory and neuropathic pain. When siRNA targeting Ift88, a primary cilium-specific intraflagellar transport (IFT) protein required for ciliary integrity, was administered by intrathecal injection, in the rat, it resulted in loss of Ift88 mRNA in DRG, and primary cilia in neuronal cell bodies, which was associated with an increase in mechanical nociceptive threshold, and abrogation of hyperalgesia induced by the pronociceptive inflammatory mediator, prostaglandin E2, and painful peripheral neuropathy induced by a neurotoxic chemotherapy drug, paclitaxel. To provide further support for the role of the primary cilium in nociceptor function we also administered siRNA for another IFT protein, Ift52. Ift52 siRNA results in loss of Ift52 in DRG and abrogates paclitaxel-induced painful peripheral neuropathy. Attenuation of Hedgehog-induced hyperalgesia by Ift88 knockdown supports a role for the primary cilium in the hyperalgesia induced by Hedgehog, and attenuation of paclitaxel chemotherapy-induced neuropathy (CIPN) by cyclopamine, which attenuates Hedgehog signaling, suggests a role of Hedgehog in CIPN. Our findings support a role of nociceptor primary cilia in the control of mechanical nociceptive threshold and in inflammatory and neuropathic pain, the latter, at least in part, Hedgehog dependent.

3.
Mol Pain ; 20: 17448069241230419, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38246917

RESUMEN

In vivo analysis of protein function in nociceptor subpopulations using antisense oligonucleotides and short interfering RNAs is limited by their non-selective cellular uptake. To address the need for selective transfection methods, we covalently linked isolectin B4 (IB4) to streptavidin and analyzed whether it could be used to study protein function in IB4(+)-nociceptors. Rats treated intrathecally with IB4-conjugated streptavidin complexed with biotinylated antisense oligonucleotides for protein kinase C epsilon (PKCε) mRNA were found to have: (a) less PKCε in dorsal root ganglia (DRG), (b) reduced PKCε expression in IB4(+) but not IB4(-) DRG neurons, and (c) fewer transcripts of the PKCε gene in the DRG. This knockdown in PKCε expression in IB4(+) DRG neurons is sufficient to reverse hyperalgesic priming, a rodent model of chronic pain that is dependent on PKCε in IB4(+)-nociceptors. These results establish that IB4-streptavidin can be used to study protein function in a defined subpopulation of nociceptive C-fiber afferents.


Asunto(s)
Lectinas , Nociceptores , Ratas , Animales , Lectinas/metabolismo , Nociceptores/metabolismo , Estreptavidina/metabolismo , Ratas Sprague-Dawley , Fibras Nerviosas Amielínicas/metabolismo , Oligonucleótidos Antisentido/metabolismo , Ganglios Espinales/metabolismo
4.
Brain ; 147(3): 1025-1042, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-37787114

RESUMEN

Progress in the development of effective chemotherapy is producing a growing population of patients with acute and chronic painful chemotherapy-induced peripheral neuropathy (CIPN), a serious treatment-limiting side effect for which there is currently no US Food and Drug Administration-approved treatment. CIPNs induced by diverse classes of chemotherapy drugs have remarkably similar clinical presentations, leading to the suggestion they share underlying mechanisms. Sensory neurons share with immune cells the ability to detect damage associated molecular patterns (DAMPs), molecules produced by diverse cell types in response to cellular stress and injury, including by chemotherapy drugs. DAMPs, in turn, are ligands for pattern recognition receptors (PRRs), several of which are found on sensory neurons, as well as satellite cells, and cells of the immune system. In the present experiments, we evaluated the role of two PRRs, TLR4 and RAGE, present in dorsal root ganglion (DRG), in CIPN. Antisense (AS)-oligodeoxynucleotides (ODN) against TLR4 and RAGE mRNA were administered intrathecally before ('prevention protocol') or 3 days after ('reversal protocol') the last administration of each of three chemotherapy drugs that treat cancer by different mechanisms (oxaliplatin, paclitaxel and bortezomib). TLR4 and RAGE AS-ODN prevented the development of CIPN induced by all three chemotherapy drugs. In the reversal protocol, however, while TLR4 AS-ODN completely reversed oxaliplatin- and paclitaxel-induced CIPN, in rats with bortezomib-induced CIPN it only produced a temporary attenuation. RAGE AS-ODN, in contrast, reversed CIPN induced by all three chemotherapy drugs. When a TLR4 antagonist was administered intradermally to the peripheral nociceptor terminal, it did not affect CIPN induced by any of the chemotherapy drugs. However, when administered intrathecally, to the central terminal, it attenuated hyperalgesia induced by all three chemotherapy drugs, compatible with a role of TLR4 in neurotransmission at the central terminal but not sensory transduction at the peripheral terminal. Finally, since it has been established that cultured DRG neurons can be used to study direct effects of chemotherapy on nociceptors, we also evaluated the role of TLR4 in CIPN at the cellular level, using patch-clamp electrophysiology in DRG neurons cultured from control and chemotherapy-treated rats. We found that increased excitability of small-diameter DRG neurons induced by in vivo and in vitro exposure to oxaliplatin is TLR4-dependent. Our findings suggest that in addition to the established contribution of PRR-dependent neuroimmune mechanisms, PRRs in DRG cells also have an important role in CIPN.


Asunto(s)
Antineoplásicos , Neuralgia , Humanos , Estados Unidos , Animales , Ratas , Bortezomib , Oxaliplatino/toxicidad , Receptor Toll-Like 4 , Neuralgia/inducido químicamente , Células Receptoras Sensoriales , Oligodesoxirribonucleótidos , Paclitaxel , Antineoplásicos/toxicidad
5.
bioRxiv ; 2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38234719

RESUMEN

The primary cilium, a 1-3 µm long hair-like structure protruding from the surface of almost all cells in the vertebrate body, is critical for neuronal development and also functions in the adult. As the migratory neural crest settles into dorsal root ganglia (DRG) sensory neurons elaborate a single primary cilium at their soma that is maintained into adult stages. While it is not known if primary cilia are expressed in nociceptors, or their potential function in the mature DRG neuron, recent studies have shown a role for Hedgehog, whose signaling demonstrates a dependence on primary cilia, in nociceptor sensitization. Here we report the expression of primary cilia in rat and mouse nociceptors, where they modulate mechanical nociceptive threshold, and contribute to inflammatory and neuropathic pain. When siRNA targeting Ift88 , a primary cilium-specific intra-flagellar transport (IFT) protein required for ciliary integrity, was administered by intrathecal injection, in the rat, it resulted in loss of Ift88 mRNA in DRG, and primary cilia in neuronal cell bodies, which was associated with an increase in mechanical nociceptive threshold, and abrogation of hyperalgesia induced by the pronociceptive inflammatory mediator, prostaglandin E 2 , and painful peripheral neuropathy induced by a neurotoxic chemotherapy drug, paclitaxel. To provide further support for the role of the primary cilium in nociceptor function we also administered siRNA for another IFT protein, Ift 52. Ift 52 siRNA results in loss of Ift 52 in DRG and abrogates paclitaxel-induced painful peripheral neuropathy. Attenuation of Hedgehog-induced hyperalgesia by Ift88 knockdown supports a role for the primary cilium in the hyperalgesia induced by Hedgehog, and attenuation of paclitaxel chemotherapy-induced neuropathy (CIPN) by cyclopamine, which attenuates Hedgehog signaling, suggests a role of Hedgehog in CIPN. Our findings support a role of nociceptor primary cilia in the control of mechanical nociceptive threshold and in inflammatory and neuropathic pain, the latter, at least in part, Hedgehog dependent.

6.
J Pain ; 22(7): 806-816, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33636374

RESUMEN

Adult rats previously submitted to neonatal limited bedding (NLB), a model of early-life stress, display muscle mechanical hyperalgesia and nociceptor hyperexcitability, the underlying mechanism for which is unknown. Since voltage-gated sodium channel subtype 7 (NaV1.7) contributes to mechanical hyperalgesia in several preclinical pain models and is critical for nociceptor excitability, we explored its role in the muscle hyperalgesia exhibited by adult NLB rats. Western blot analyses demonstrated increased NaV1.7 protein expression in L4-L5 dorsal root ganglia (DRG) from adult NLB rats, and antisense oligodeoxynucleotide (AS ODN) targeting NaV1.7 alpha subunit mRNA attenuated the expression of NaV1.7 in DRG extracts. While this AS ODN did not affect nociceptive threshold in normal rats it significantly attenuated hyperalgesia in NLB rats. The selective NaV1.7 activator OD1 produced dose-dependent mechanical hyperalgesia that was enhanced in NLB rats, whereas the NaV1.7 blocker ProTx-II prevented OD1-induced hyperalgesia in control rats and ongoing hyperalgesia in NLB rats. AS ODN knockdown of extracellular signal-regulated kinase 1/2, which enhances NaV1.7 function, also inhibited mechanical hyperalgesia in NLB rats. Our results support the hypothesis that overexpression of NaV1.7 in muscle nociceptors play a role in chronic muscle pain induced by early-life stress, suggesting that NaV1.7 is a target for the treatment of chronic muscle pain. PERSPECTIVE: We demonstrate that early-life adversity, induced by exposure to inconsistent maternal care, produces chronic muscle hyperalgesia, which depends, at least in part, on increased expression of NaV1.7 in nociceptors.


Asunto(s)
Dolor Crónico , Hiperalgesia , Mialgia , Canal de Sodio Activado por Voltaje NAV1.7 , Nociceptores , Estrés Psicológico , Animales , Femenino , Masculino , Ratas , Animales Recién Nacidos , Dolor Crónico/etiología , Dolor Crónico/metabolismo , Modelos Animales de Enfermedad , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Sistema de Señalización de MAP Quinasas , Mialgia/etiología , Mialgia/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Nocicepción , Nociceptores/metabolismo , Umbral del Dolor , Ratas Sprague-Dawley
7.
J Pain ; 22(5): 498-508, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33232830

RESUMEN

TACAN (Tmem120A), a mechanotransducing ion channel highly expressed in a subset of nociceptors, has recently been shown to contribute to detection of noxious mechanical stimulation. In the present study we evaluated its role in sensitization to mechanical stimuli associated with preclinical models of inflammatory and chemotherapy-induced neuropathic pain (CIPN). Intrathecal administration of an oligodeoxynucleotide antisense (AS-ODN) to TACAN mRNA attenuated TACAN protein expression in rat dorsal root ganglia (DRG). While TACAN AS-ODN produced only a modest increase in mechanical nociceptive threshold, it markedly reduced mechanical hyperalgesia produced by intradermal administration of prostaglandin E2, tumor necrosis factor alpha, and low molecular weight hyaluronan, and systemic administration of lipopolysaccharide, compatible with a prominent role of TACAN in mechanical hyperalgesia produced by inflammation. In contrast, TACAN AS-ODN had no effect on mechanical hyperalgesia associated with CIPN produced by oxaliplatin or paclitaxel. Our results provide evidence that TACAN plays a role in mechanical hyperalgesia induced by pronociceptive inflammatory mediators, but not CIPN, compatible with multiple mechanisms mediating mechanical nociception, and sensitization to mechanical stimuli in preclinical models of inflammatory versus CIPN. PERSPECTIVE: We evaluated the role of TACAN, a mechanotransducing ion channel in nociceptors, in preclinical models of inflammatory and CIPN. Attenuation of TACAN expression reduced hyperalgesia produced by inflammatory mediators but had not chemotherapeutic agents. Our findings support the presence of multiple mechanotransducers in nociceptors.


Asunto(s)
Antineoplásicos/efectos adversos , Ganglios Espinales/fisiología , Hiperalgesia/fisiopatología , Inflamación/complicaciones , Canales Iónicos/farmacología , Mecanotransducción Celular/fisiología , Neuralgia/etiología , Neuralgia/fisiopatología , Umbral del Dolor/fisiología , Animales , Modelos Animales de Enfermedad , Masculino , Neuralgia/inducido químicamente , Oxaliplatino/efectos adversos , Paclitaxel/efectos adversos , Ratas , Ratas Sprague-Dawley
8.
J Neurosci ; 40(34): 6477-6488, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32665406

RESUMEN

We evaluated the mechanism by which high-molecular-weight hyaluronan (HMWH) attenuates nociceptor sensitization, in the setting of inflammation. HMWH attenuated mechanical hyperalgesia induced by the inflammatory mediator prostaglandin E2 (PGE2) in male and female rats. Intrathecal administration of an oligodeoxynucleotide antisense (AS-ODN) to mRNA for cluster of differentiation 44 (CD44), the cognate hyaluronan receptor, and intradermal administration of A5G27, a CD44 receptor antagonist, both attenuated antihyperalgesia induced by HMWH. In male rats, HMWH also signals via Toll-like receptor 4 (TLR4), and AS-ODN for TLR4 mRNA administered intrathecally, attenuated HMWH-induced antihyperalgesia. Since HMWH signaling is dependent on CD44 clustering in lipid rafts, we pretreated animals with methyl-ß-cyclodextrin (MßCD), which disrupts lipid rafts. MßCD markedly attenuated HMWH-induced antihyperalgesia. Inhibitors for components of intracellular signaling pathways activated by CD44, including phospholipase C and phosphoinositide 3-kinase (PI3K), also attenuated HMWH-induced antihyperalgesia. Furthermore, in vitro application of HMWH attenuated PGE2-induced sensitization of tetrodotoxin-resistant sodium current, in small-diameter dorsal root ganglion neurons, an effect that was attenuated by a PI3K inhibitor. Our results indicate a central role of CD44 signaling in HMWH-induced antihyperalgesia and suggest novel therapeutic targets, downstream of CD44, for the treatment of pain generated by nociceptor sensitization.SIGNIFICANCE STATEMENT High-molecular-weight-hyaluronan (HMWH) is used to treat osteoarthritis and other pain syndromes. In this study we demonstrate that attenuation of inflammatory hyperalgesia by HMWH is mediated by its action at cluster of differentiation 44 (CD44) and activation of its downstream signaling pathways, including RhoGTPases (RhoA and Rac1), phospholipases (phospholipases Cε and Cγ1), and phosphoinositide 3-kinase, in nociceptors. These findings contribute to our understanding of the antihyperalgesic effect of HMWH and support the hypothesis that CD44 and its downstream signaling pathways represent novel therapeutic targets for the treatment of inflammatory pain.


Asunto(s)
Ácido Hialurónico/metabolismo , Hiperalgesia/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Dinoprostona/administración & dosificación , Femenino , Ganglios Espinales/metabolismo , Receptores de Hialuranos/metabolismo , Hiperalgesia/inducido químicamente , Masculino , Nocicepción/fisiología , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo
9.
J Pain ; 21(3-4): 506-512, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31562993

RESUMEN

Occupational exposure to mechanical vibration can produce the hand-arm vibration syndrome (HAVS), whose most disabling symptom is persistent muscle pain. Unfortunately, the pathophysiology of HAVS pain is still poorly understood, precluding the development of mechanism-based therapies. Since interleukin 33 (IL-33) is essential for inflammation and recovery that follows skeletal muscle injury, we explored its role in muscle pain in a model of HAVS, in adult male rats. Concomitant to mechanical hyperalgesia, an increase in IL-33 in the ipsilateral gastrocnemius muscle was observed 24 hours after vibration. A similar hyperalgesia was produced by intramuscular injection of recombinant rat IL-33 (rrIL-33, 10-300 ng). Intrathecal administration of an oligodeoxynucleotide antisense to IL-33R/ST2 mRNA decreased the expression of ST2 in DRG and attenuated both rrIL-33 and vibration-induced mechanical hyperalgesia. Together these data support the suggestion that IL-33 plays a central role in vibration-induced muscle pain by action, at least in part, on skeletal muscle nociceptors. PERSPECTIVE: Our findings provide evidence of the contribution of IL-33, acting on its canonical receptor, in nociceptors, to muscle pain induced by ergonomic vibration. This suggests that targeting IL-33/ST2 signaling may be a useful strategy for the treatment of muscle pain in HAVS.


Asunto(s)
Síndrome por Vibración de la Mano y el Brazo/metabolismo , Síndrome por Vibración de la Mano y el Brazo/fisiopatología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/administración & dosificación , Interleucina-33/metabolismo , Mialgia/metabolismo , Nociceptores/fisiología , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inyecciones Intramusculares , Masculino , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Vibración/efectos adversos
11.
Pain ; 160(8): 1876-1882, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31335655

RESUMEN

Chronic muscle pain is a prominent symptom of the hand-arm vibration syndrome (HAVS), an occupational disease induced by exposure to vibrating power tools, but the underlying mechanism remains unknown. We evaluated the hypothesis that vibration induces an interleukin 6 (IL-6)-mediated downregulation of the potassium voltage-gated channel subfamily A member 4 (KV1.4) in nociceptors leading to muscle pain. Adult male rats were submitted to a protocol of mechanical vibration of the right hind limb. Twenty-four hours after vibration, muscle hyperalgesia was observed, concomitant to increased levels of IL-6 in the gastrocnemius muscle and decreased expression of KV1.4 in the dorsal root ganglia. Local injection of neutralizing antibodies against IL-6 attenuated the muscle hyperalgesia induced by vibration, whereas antisense knockdown of this channel in the dorsal root ganglia mimicked the muscle hyperalgesia observed in the model of HAVS. Finally, knockdown of the IL-6 receptor signaling subunit glycoprotein 130 (gp130) attenuated both vibration-induced muscle hyperalgesia and downregulation of KV1.4. These results support the hypothesis that IL-6 plays a central role in the induction of muscle pain in HAVS. This likely occurs through intracellular signaling downstream to the IL-6 receptor subunit gp130, which decreases the expression of KV1.4 in nociceptors.


Asunto(s)
Ganglios Espinales/metabolismo , Síndrome por Vibración de la Mano y el Brazo/metabolismo , Hiperalgesia/metabolismo , Interleucina-6/metabolismo , Canal de Potasio Kv1.4/metabolismo , Músculo Esquelético/metabolismo , Dolor Musculoesquelético/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Síndrome por Vibración de la Mano y el Brazo/genética , Interleucina-6/genética , Canal de Potasio Kv1.4/genética , Masculino , Dolor Musculoesquelético/genética , Ratas , Ratas Sprague-Dawley , Vibración
12.
J Neurosci ; 39(33): 6414-6424, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31209174

RESUMEN

In addition to analgesia, opioids produce opioid-induced hyperalgesia (OIH) and neuroplasticity characterized by prolongation of inflammatory-mediator-induced hyperalgesia (hyperalgesic priming). We evaluated the hypothesis that hyperalgesia and priming induced by opioids are mediated by similar nociceptor mechanisms. In male rats, we first evaluated the role of nociceptor Toll-like receptor 4 (TLR4) in OIH and priming induced by systemic low-dose morphine (LDM, 0.03 mg/kg). Intrathecal oligodeoxynucleotide antisense to TLR4 mRNA (TLR4 AS-ODN) prevented OIH and prolongation of prostaglandin E2 hyperalgesia (priming) induced by LDM. In contrast, high-dose morphine (HDM, 3 mg/kg) increased nociceptive threshold (analgesia) and induced priming, neither of which was attenuated by TLR4 AS-ODN. Protein kinase C ε (PKCε) AS-ODN also prevented LDM-induced hyperalgesia and priming, whereas analgesia and priming induced by HDM were unaffected. Treatment with isolectin B4 (IB4)-saporin or SSP-saporin (which deplete IB4+ and peptidergic nociceptors, respectively), or their combination, prevented systemic LDM-induced hyperalgesia, but not priming. HDM-induced priming, but not analgesia, was markedly attenuated in both saporin-treated groups. In conclusion, whereas OIH and priming induced by LDM share receptor and second messenger mechanisms in common, action at TLR4 and signaling via PKCε, HDM-induced analgesia, and priming are neither TLR4 nor PKCε dependent. OIH produced by LDM is mediated by both IB4+ and peptidergic nociceptors, whereas priming is not dependent on the same population. In contrast, priming induced by HDM is mediated by both IB4+ and peptidergic nociceptors. Implications for the use of low-dose opioids combined with nonopioid analgesics and in the treatment of opioid use disorder are discussed.SIGNIFICANCE STATEMENT Opioid-induced hyperalgesia (OIH) and priming are common side effects of opioid agonists such as morphine, which acts at µ-opioid receptors. We demonstrate that OIH and priming induced by systemic low-dose morphine (LDM) share action at Toll-like receptor 4 (TLR4) and signaling via protein kinase C ε (PKCε) in common, whereas systemic high-dose morphine (HDM)-induced analgesia and priming are neither TLR4 nor PKCε dependent. OIH produced by systemic LDM is mediated by isolectin B4-positive (IB4+) and peptidergic nociceptors, whereas priming is dependent on a different class of nociceptors. Priming induced by systemic HDM is, however, mediated by both IB4+ and peptidergic nociceptors. Our findings may provide useful information for the use of low-dose opioids combined with nonopioid analgesics to treat pain and opioid use disorders.


Asunto(s)
Analgésicos Opioides/farmacología , Hiperalgesia/metabolismo , Morfina/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Hiperalgesia/inducido químicamente , Masculino , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
13.
Neuroscience ; 398: 64-75, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529265

RESUMEN

We investigated the dose dependence of the role of nociceptors in opioid-induced side-effects, hyperalgesia and pain chronification, in the rat. Systemic morphine produced a dose-dependent biphasic change in mechanical nociceptive threshold. At lower doses (0.003-0.03 mg/kg, s.c.) morphine induced mechanical hyperalgesia, while higher doses (1-10 mg/kg, s.c.) induced analgesia. Intrathecal (i.t.) oligodeoxynucleotide (ODN) antisense to mu-opioid receptor (MOR) mRNA, attenuated both hyperalgesia and analgesia. 5 days after systemic morphine (0.03-10 mg/kg s.c.), mechanical hyperalgesia produced by intradermal (i.d.) prostaglandin E2 (PGE2) was prolonged, indicating hyperalgesic priming at the peripheral terminal of the nociceptor. The hyperalgesia induced by i.t. PGE2 (400 ng/10 µl), in groups that received 0.03 (that induced hyperalgesia) or 3 mg/kg (that induced analgesia) morphine, was also prolonged, indicating priming at the central terminal of the nociceptor. The prolongation of the hyperalgesia induced by i.d. or i.t. PGE2, in rats previously treated with either a hyperalgesic (0.03 mg/kg, s.c.) or analgesic (3 mg/kg, s.c.) dose, was reversed by i.d. or i.t. injection of the protein translation inhibitor cordycepin (1 µg), indicative of Type I priming at both terminals. Although pretreatment with MOR antisense had no effect on priming induced by 0.03 mg/kg morphine, it completely prevented priming by 3 mg/kg morphine, in both terminals. Thus, the induction of hyperalgesia, but not priming, by low-dose morphine, is MOR-dependent. In contrast, induction of both hyperalgesia and priming by high-dose morphine is MOR-dependent. The receptor at which low-dose morphine acts to produce priming remains to be established.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Nociceptores/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Masculino , Plasticidad Neuronal/fisiología , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/fisiopatología , Nociceptores/fisiología , Umbral del Dolor/fisiología , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo
14.
Pain ; 158(8): 1481-1488, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28628078

RESUMEN

Delayed-onset muscle soreness is typically observed after strenuous or unaccustomed eccentric exercise. Soon after recovery, blunted muscle soreness is observed on repeated eccentric exercise, a phenomenon known as repeated bout effect (RBE). Although regular physical activity decreases muscle hyperalgesia, likely because of increased production of the anti-inflammatory cytokine interleukin-10 (IL-10) in the skeletal muscle, whether IL-10 also contributes to the antinociceptive effect of RBE is unknown. Furthermore, whether IL-10 attenuates muscle hyperalgesia by acting on muscle nociceptors remains to be established. Here, we explored the hypothesis that blunted muscle nociception observed in RBE depends on a local effect of IL-10, acting on IL-10 receptor 1 (IL-10R1) expressed by muscle nociceptors. Results show that after a second bout of eccentric exercise, rats exhibited decreased muscle hyperalgesia, indicative of RBE, and increased expression of IL-10 in the exercised gastrocnemius muscle. Although knockdown of IL-10R1 protein in nociceptors innervating the gastrocnemius muscle by intrathecal antisense oligodeoxynucleotide did not change nociceptive threshold in naive rats, it unveiled latent muscle hyperalgesia in rats submitted to eccentric exercise 12 days ago. Furthermore, antisense also prevented the reduction of muscle hyperalgesia observed after a second bout of eccentric exercise. These data indicate that recovery of nociceptive threshold after eccentric exercise and RBE-induced analgesia depend on a local effect of IL-10, acting on its canonical receptor in muscle nociceptors.


Asunto(s)
Hiperalgesia/metabolismo , Subunidad alfa del Receptor de Interleucina-10/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Interleucina-10/metabolismo , Masculino , Nociceptores/metabolismo , Manejo del Dolor/métodos , Ratas Sprague-Dawley
15.
J Pain ; 17(4): 444-50, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26721612

RESUMEN

UNLABELLED: We hypothesized that changes in the expression of voltage-gated potassium channel (Kv) 4.3 contribute to the mechanical hyperalgesia induced by vibration injury, in a rodent model for hand-arm vibration syndrome in humans. Here we show that the exposure of the gastrocnemius muscle to vibration injury induces muscle hyperalgesia that is accompanied by a significant downregulation of Kv4.3 in affected sensory nerve fibers in dorsal root ganglia. We additionally show that the intrathecal administration of antisense oligonucleotides for Kv4.3 messenger RNA itself induces muscle hyperalgesia in the rat. Our results suggest that attenuation in the expression of Kv4.3 may contribute to neuropathic pain in people affected by hand-arm vibration syndrome. PERSPECTIVE: Our findings establish Kv4.3 as a potential molecular target for the treatment of hand-arm vibration syndrome.


Asunto(s)
Regulación hacia Abajo/fisiología , Mialgia/etiología , Mialgia/metabolismo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Vibración/efectos adversos , Animales , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Glicoproteínas/metabolismo , Hiperalgesia/metabolismo , Lectinas/metabolismo , Masculino , Umbral del Dolor/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Versicanos
16.
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
17.
J Neurochem ; 134(1): 147-55, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25845936

RESUMEN

A subpopulation of nociceptors, the glial cell line-derived neurotrophic factor (GDNF)-dependent, non-peptidergic C-fibers, expresses a cell-surface glycoconjugate that can be selectively labeled with isolectin B4 (IB4 ), a homotetrameric plant lectin from Griffonia simplicifolia. We show that versican is an IB4 -binding molecule in rat dorsal root ganglion neurons. Using reverse transcriptase polymerase chain reaction (RT-PCR), in situ hybridization and immunofluorescence experiments on rat lumbar dorsal root ganglion, we provide the first demonstration that versican is produced by neurons. In addition, by probing Western blots with splice variant-specific antibodies we show that the IB4 -binding versican contains only the glycosaminoglycan alpha domain. Our data support V2 as the versican isoform that renders this subpopulation of nociceptors IB4 -positive (+). A subset of nociceptors, the GDNF-dependent non-peptidergic C-fibers can be characterized by its reactivity for isolectin B4 (IB4), a plant lectin from Griffonia simplicifolia. We have previously demonstrated that versican V2 binds IB4 in a Ca2 + -dependent manner. However, given that versican is thought to be the product of glial cells, it was questionable whether versican V2 can be accountable for the IB4-reactivity of this subset of nociceptors. The results presented here prove - for the first time - a neuronal origin of versican and suggest that versican V2 is the molecule that renders GDNF-dependent non-peptidergic C-fibers IB4-positive.


Asunto(s)
Glicoproteínas/metabolismo , Lectinas/metabolismo , Fibras Nerviosas Amielínicas/metabolismo , Neuronas/metabolismo , Nociceptores/metabolismo , Versicanos/metabolismo , Animales , Ganglios Espinales/metabolismo , Glicoproteínas/análisis , Lectinas/análisis , Masculino , Fibras Nerviosas Amielínicas/química , Neuronas/química , Nociceptores/química , Ratas , Ratas Sprague-Dawley , Versicanos/análisis
18.
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
19.
Pain ; 155(12): 2680-2686, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25280432

RESUMEN

Endometriosis, the most common cause of chronic pelvic pain, is an estrogen-dependent disease in which classic estrogen receptors (ERα, ERß) play an important role. Although recent evidence suggests that the novel G protein-coupled estrogen receptor (GPR30) also plays a key role in the progression of endometriosis, whether it is also involved in endometriosis pain is still unknown. Here we tested the hypothesis that GPR30 expressed by nociceptors contributes to endometriosis pain. Intramuscular injection of the GPR30 agonists raloxifene or 17ß-estradiol produced a fast-onset, persistent, mechanical hyperalgesia at the site of the injection. Intrathecal antisense (AS) oligodeoxynucleotides (ODN), but not mismatch (MM) ODN, targeting mRNA for GPR30 markedly inhibited its protein expression in nociceptors and attenuated the mechanical hyperalgesia induced by local raloxifene or 17ß-estradiol. Pretreatment with the GPR30 antagonist G-36 also inhibited the hyperalgesia induced by raloxifene or 17ß-estradiol in naive control rats. Surgical implant of autologous uterine tissue onto the gastrocnemius muscle, which induces endometriosis-like lesions, produced local mechanical hyperalgesia. Intrathecal AS, but not MM, ODN targeting GPR30 mRNA reversibly inhibited the mechanical hyperalgesia at the site of endometriotic lesions. Finally, intralesional injection of the GPR30 antagonist G-36 also inhibited the mechanical hyperalgesia at the site of ectopic uterine tissue. We conclude that local GPR30 agonists produce persistent mechanical hyperalgesia in naive female rats, whereas local GPR30 antagonists inhibit mechanical hyperalgesia in a model of endometriosis pain. Thus, GPR30 expressed by nociceptors innervating ectopic uterine lesions might play a major role in endometriosis pain.


Asunto(s)
Endometriosis/complicaciones , Regulación de la Expresión Génica/fisiología , Nociceptores/metabolismo , Dolor Pélvico/etiología , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endometriosis/inducido químicamente , Estradiol/efectos adversos , Antagonistas de Estrógenos/efectos adversos , Femenino , Ganglios Espinales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/inducido químicamente , Nociceptores/efectos de los fármacos , Oligonucleótidos Antisentido , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Proteína Quinasa C-epsilon/metabolismo , ARN Mensajero/metabolismo , Clorhidrato de Raloxifeno/efectos adversos , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
20.
J Pain ; 15(3): 312-20, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24407022

RESUMEN

UNLABELLED: Hyperalgesic priming is a model of the transition from acute to chronic pain, in which previous activation of cell surface receptors or direct activation of protein kinase C epsilon markedly prolongs mechanical hyperalgesia induced by pronociceptive cytokines. We recently demonstrated a role of peripheral protein translation, alpha-calmodulin-dependent protein kinase II (αCaMKII) activation, and the ryanodine receptor in the induction of hyperalgesic priming. In the present study, we tested if they also mediate the prolonged phase of prostaglandin E2-induced hyperalgesia. We found that inhibition of αCaMKII and local protein translation eliminates the prolonged phase of prostaglandin E2 hyperalgesia. Although priming induced by receptor agonists or direct activation of protein kinase C epsilon occurs in male but not female rats, activation of αCaMKII and the ryanodine receptor also produces priming in females. As in males, the prolonged phase of prostaglandin E2-induced hyperalgesia in female rats is also protein kinase C epsilon-, αCaMKII-, and protein translation-dependent. In addition, in both male and female primed rats, the prolonged prostaglandin E2-induced hyperalgesia was significantly attenuated by inhibition of MEK/ERK. On the basis of these data, we suggest that the mechanisms previously shown to be involved in the induction of the neuroplastic state of hyperalgesic priming also mediate the prolongation of hyperalgesia. PERSPECTIVES: The data provided by this study suggest that direct intervention on specific targets may help to alleviate the expression of chronic hyperalgesic conditions.


Asunto(s)
Dolor Crónico/fisiopatología , Hiperalgesia/fisiopatología , Plasticidad Neuronal/fisiología , Sistemas de Mensajero Secundario/fisiología , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/metabolismo , Dinoprostona , Femenino , Hiperalgesia/inducido químicamente , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Biosíntesis de Proteínas , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Caracteres Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...