Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 4696, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32929092

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Haematologica ; 103(7): 1124-1135, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29545351

RESUMEN

Sickle cell disease is associated with acute painful episodes and chronic intractable pain. Endothelin-1, a known pain inducer, is elevated in the blood plasma of both sickle cell patients and mouse models of sickle cell disease. We show here that the levels of endothelin-1 and its endothelin type A receptor are increased in the dorsal root ganglia of a mouse model of sickle cell disease. Pharmacologic inhibition or neuron-specific knockdown of endothelin type A receptors in primary sensory neurons of dorsal root ganglia alleviated basal and post-hypoxia evoked pain hypersensitivities in sickle cell mice. Mechanistically, endothelin type A receptors contribute to sickle cell disease-associated pain likely through the activation of NF-κB-induced Nav1.8 channel upregulation in primary sensory neurons of sickle cell mice. Our findings suggest that endothelin type A receptor is a potential target for the management of sickle cell disease-associated pain, although this expectation needs to be further verified in clinical settings.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Dolor/etiología , Receptor de Endotelina A/genética , Anemia de Células Falciformes/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Endotelina-1/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Hiperalgesia/diagnóstico , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Dolor/diagnóstico , Dolor/metabolismo , Células del Asta Posterior/metabolismo , Receptor de Endotelina A/metabolismo
3.
Nat Commun ; 8: 14712, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28270689

RESUMEN

Nerve injury induces changes in gene transcription in dorsal root ganglion (DRG) neurons, which may contribute to nerve injury-induced neuropathic pain. DNA methylation represses gene expression. Here, we report that peripheral nerve injury increases expression of the DNA methyltransferase DNMT3a in the injured DRG neurons via the activation of the transcription factor octamer transcription factor 1. Blocking this increase prevents nerve injury-induced methylation of the voltage-dependent potassium (Kv) channel subunit Kcna2 promoter region and rescues Kcna2 expression in the injured DRG and attenuates neuropathic pain. Conversely, in the absence of nerve injury, mimicking this increase reduces the Kcna2 promoter activity, diminishes Kcna2 expression, decreases Kv current, increases excitability in DRG neurons and leads to spinal cord central sensitization and neuropathic pain symptoms. These findings suggest that DNMT3a may contribute to neuropathic pain by repressing Kcna2 expression in the DRG.


Asunto(s)
Sensibilización del Sistema Nervioso Central/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Canal de Potasio Kv.1.2/genética , Neuralgia/genética , Neuronas Aferentes/metabolismo , Traumatismos de los Nervios Periféricos/genética , Animales , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Regulación de la Expresión Génica , Canal de Potasio Kv.1.2/metabolismo , Ligadura , Masculino , Factor 1 de Transcripción de Unión a Octámeros/genética , Ratas , Nervios Espinales/lesiones
4.
Sci Rep ; 6: 37704, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27874088

RESUMEN

Nerve injury-induced downregulation of voltage-gated potassium channel subunit Kcna2 in the dorsal root ganglion (DRG) is critical for DRG neuronal excitability and neuropathic pain genesis. However, how nerve injury causes this downregulation is still elusive. Euchromatic histone-lysine N-methyltransferase 2, also known as G9a, methylates histone H3 on lysine residue 9 to predominantly produce a dynamic histone dimethylation, resulting in condensed chromatin and gene transcriptional repression. We showed here that blocking nerve injury-induced increase in G9a rescued Kcna2 mRNA and protein expression in the axotomized DRG and attenuated the development of nerve injury-induced pain hypersensitivity. Mimicking this increase decreased Kcna2 mRNA and protein expression, reduced Kv current, and increased excitability in the DRG neurons and led to spinal cord central sensitization and neuropathic pain-like symptoms. G9a mRNA is co-localized with Kcna2 mRNA in the DRG neurons. These findings indicate that G9a contributes to neuropathic pain development through epigenetic silencing of Kcna2 in the axotomized DRG.


Asunto(s)
Regulación hacia Abajo/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Canal de Potasio Kv.1.2/genética , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Nervios Espinales/lesiones , Potenciales de Acción , Animales , Axotomía , Células Cultivadas , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Histonas/metabolismo , Hipersensibilidad/patología , Hipersensibilidad/fisiopatología , Activación del Canal Iónico , Canal de Potasio Kv.1.2/metabolismo , Ligadura , Lisina/metabolismo , Masculino , Metilación , Ratones Endogámicos C57BL , Neuralgia/patología , Neuralgia/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Nervios Espinales/patología , Nervios Espinales/fisiopatología
5.
Anesthesiology ; 125(4): 765-78, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27483126

RESUMEN

BACKGROUND: Peripheral nerve injury-induced gene alterations in the dorsal root ganglion (DRG) and spinal cord likely participate in neuropathic pain genesis. Histone methylation gates gene expression. Whether the suppressor of variegation 3-9 homolog 1 (SUV39H1), a histone methyltransferase, contributes to nerve injury-induced nociceptive hypersensitivity is unknown. METHODS: Quantitative real-time reverse transcription polymerase chain reaction analysis, Western blot analysis, or immunohistochemistry were carried out to examine the expression of SUV39H1 mRNA and protein in rat DRG and dorsal horn and its colocalization with DRG µ-opioid receptor (MOR). The effects of a SUV39H1 inhibitor (chaetocin) or SUV39H1 siRNA on fifth lumbar spinal nerve ligation (SNL)-induced DRG MOR down-regulation and nociceptive hypersensitivity were examined. RESULTS: SUV39H1 was detected in neuronal nuclei of the DRG and dorsal horn. It was distributed predominantly in small DRG neurons, in which it coexpressed with MOR. The level of SUV39H1 protein in both injured DRG and ipsilateral fifth lumbar dorsal horn was time dependently increased after SNL. SNL also produced an increase in the amount of SUV39H1 mRNA in the injured DRG (n = 6/time point). Intrathecal chaetocin or SUV39H1 siRNA as well as DRG or intraspinal microinjection of SUV39H1 siRNA impaired SNL-induced allodynia and hyperalgesia (n = 5/group/treatment). DRG microinjection of SUV39H1 siRNA also restored SNL-induced DRG MOR down-regulation (n = 6/group). CONCLUSIONS: The findings of this study suggest that SUV39H1 contributes to nerve injury-induced allodynia and hyperalgesia through gating MOR expression in the injured DRG. SUV39H1 may be a potential target for the therapeutic treatment of nerve injury-induced nociceptive hypersensitivity.


Asunto(s)
Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Metiltransferasas/metabolismo , Neuralgia/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas Represoras/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Hiperalgesia/genética , Inmunohistoquímica , Masculino , Metiltransferasas/genética , Neuralgia/genética , Traumatismos de los Nervios Periféricos/genética , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Transl Perioper Pain Med ; 1(3): 22-33, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27500183

RESUMEN

The CRISPR/Cas9 system is a research hotspot in genome editing and regulation. Currently, it is used in genomic silencing and knock-in experiments as well as transcriptional activation and repression. This versatile system consists of two components: a guide RNA (gRNA) and a Cas9 nuclease. Recognition of a genomic DNA target is mediated through base pairing with a 20-base gRNA. The latter further recruits the Cas9 endonuclease protein to the target site and creates double-stranded breaks in the target DNA. Compared with traditional genome editing directed by DNA-binding protein domains, this short RNA-directed Cas9 endonuclease system is simple and easily programmable. Although this system may have off-target effects and in vivo delivery and immune challenges, researchers have employed this system in vivo to establish disease models, study specific gene functions under certain disease conditions, and correct genomic information for disease treatment. In regards to pain research, the CRISPR/Cas9 system may act as a novel tool in gene correction therapy for pain-associated hereditary diseases and may be a new approach for RNA-guided transcriptional activation or repression of pain-related genes. In addition, this system is also applied to loss-of-function mutations in pain-related genes and knockin of reporter genes or loxP tags at pain-related genomic loci. The CRISPR/Cas9 system will likely be carried out widely in both bench work and clinical settings in the pain field.

7.
Mol Pain ; 11: 73, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26626404

RESUMEN

BACKGROUND: Chronic stress has been reported to increase basal pain sensitivity and/or exacerbate existing persistent pain. However, most surgical patients have normal physiological and psychological health status such as normal pain perception before surgery although they do experience short-term stress during pre- and post-operative periods. Whether or not this short-term stress affects persistent postsurgical pain is unclear. RESULTS: In this study, we showed that pre- or post-surgical exposure to immobilization 6 h daily for three consecutive days did not change basal responses to mechanical, thermal, or cold stimuli or peak levels of incision-induced hypersensitivity to these stimuli; however, immobilization did prolong the duration of incision-induced hypersensitivity in both male and female rats. These phenomena were also observed in post-surgical exposure to forced swimming 25 min daily for 3 consecutive days. Short-term stress induced by immobilization was demonstrated by an elevation in the level of serum corticosterone, an increase in swim immobility, and a decrease in sucrose consumption. Blocking this short-term stress via intrathecal administration of a selective glucocorticoid receptor antagonist, RU38486, or bilateral adrenalectomy significantly attenuated the prolongation of incision-induced hypersensitivity to mechanical, thermal, and cold stimuli. CONCLUSION: Our results indicate that short-term stress during the pre- or post-operative period delays postoperative pain recovery although it does not affect basal pain perception. Prevention of short-term stress may facilitate patients' recovery from postoperative pain.


Asunto(s)
Percepción del Dolor/fisiología , Umbral del Dolor/fisiología , Dolor Postoperatorio/fisiopatología , Estrés Fisiológico , Estrés Psicológico , Animales , Corticosterona/sangre , Femenino , Antagonistas de Hormonas/farmacología , Humanos , Masculino , Mifepristona/farmacología , Modelos Animales , Ratas , Ratas Sprague-Dawley , Restricción Física
8.
J Pain ; 16(11): 1186-99, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342649

RESUMEN

UNLABELLED: Chronic sleep disturbance-induced stress is known to increase basal pain sensitivity. However, most surgical patients frequently report short-term sleep disturbance/deprivation during the pre- and postoperation periods and have normal pain perception presurgery. Whether this short-term sleep disturbance affects postsurgical pain is elusive. Here, we report that pre- or postexposure to rapid eye movement sleep disturbance (REMSD) for 6 hours daily for 3 consecutive days did not alter basal responses to mechanical, heat, and cold stimuli, but did delay recovery in incision-induced reductions in paw withdrawal threshold to mechanical stimulation and paw withdrawal latencies to heat and cold stimuli on the ipsilateral side of male or female rats. This short-term REMSD led to stress shown by an increase in swim immobility time, a decrease in sucrose consumption, and an increase in the level of corticosterone in serum. Blocking this stress via intrathecal RU38486 or bilateral adrenalectomy abolished REMSD-caused delay in recovery of incision-induced reductions in behavioral responses to mechanical, heat, and cold stimuli. Moreover, this short-term REMSD produced significant reductions in the levels of mu opioid receptor and kappa opioid receptor, but not Kv1.2, in the ipsilateral L4/5 spinal cord and dorsal root ganglia on day 9 after incision (but not after sham surgery). PERSPECTIVE: Our findings show that short-term sleep disturbance either pre- or postsurgery does not alter basal pain perception, but does exacerbate postsurgical pain hypersensitivity. The latter may be related to the reductions of mu and kappa opioid receptors in the spinal cord and dorsal root ganglia caused by REMSD plus incision. Prevention of short-term sleep disturbance may help recovery from postsurgical pain in patients.


Asunto(s)
Ganglios Espinales/metabolismo , Percepción del Dolor/fisiología , Dolor Postoperatorio/fisiopatología , Trastornos del Sueño-Vigilia/fisiopatología , Médula Espinal/metabolismo , Estrés Fisiológico/fisiología , Animales , Enfermedad Crónica , Corticosterona/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Ganglios Espinales/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Canal de Potasio Kv.1.2/metabolismo , Vértebras Lumbares , Masculino , Mifepristona/farmacología , Percepción del Dolor/efectos de los fármacos , Dolor Postoperatorio/tratamiento farmacológico , Ratas Sprague-Dawley , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Sueño REM/fisiología , Médula Espinal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos
9.
Transl Perioper Pain Med ; 2(2): 27-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26339682

RESUMEN

Repeated and long-term administration of opioids is often accompanied by the initiation of opioid-induced analgesic tolerance and hyperalgesia in chronic pain patients. Our previous studies showed that repeated intrathecal morphine injection activated the mammalian target of rapamycin complex 1 (mTORC1) in spinal dorsal horn neurons and that blocking this activation prevented the initiation of morphine-induced tolerance and hyperalgesia in healthy rats. However, whether spinal mTORC1 is required for morphine-induced tolerance and hyperalgesia under neuropathic pain conditions remains elusive. We here observed the effect of intrathecal infusion of rapamycin, a specific mTORC1 inhibitor, on morphine-induced tolerance and hyperalgesia in a neuropathic pain model in rats induced by the fifth lumbar spinal nerve ligation (SNL). Continuous intrathecal infusion of morphine for one week starting on day 8 post-SNL led to morphine tolerance demonstrated by morphine-induced reduction in maximal possible analgesic effect (MPAE) to tail heat stimuli and ipsilateral paw withdrawal threshold (PWT) to mechanical stimuli in SNL rats. Such reduction was attenuated by co-infusion of rapamycin. Co-infusion of rapamycin also blocked morphine tolerance demonstrated by attenuation of morphine-induced reduction in MPAE in sham rats and morphine-induced hyperalgesia demonstrated by the reverse of morphine-induced reduction in PWT on both sides of sham rats and on the contralateral side of SNL rats. The results suggest that mTORC1 inhibitors could serve as promising medications for use as adjuvants with opioids in clinical neuropathic pain management.

10.
Epigenomics ; 7(2): 235-45, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25942533

RESUMEN

Chronic pain arising from peripheral inflammation and tissue or nerve injury is a common clinical symptom. Although intensive research on the neurobiological mechanisms of chronic pain has been carried out during previous decades, this disorder is still poorly managed by current drugs such as opioids and nonsteroidal anti-inflammatory drugs. Inflammation, tissue injury and/or nerve injury-induced changes in gene expression in sensory neurons of the dorsal root ganglion, spinal cord dorsal horn and pain-associated brain regions are thought to participate in chronic pain genesis; however, how these changes occur is still elusive. Epigenetic modifications including DNA methylation and covalent histone modifications control gene expression. Recent studies have shown that peripheral noxious stimulation changes DNA methylation and histone modifications and that these changes may be related to the induction of pain hypersensitivity under chronic pain conditions. This review summarizes the current knowledge and progress in epigenetic research in chronic pain and discusses the potential role of epigenetic modifications as therapeutic antinociceptive targets in this disorder.


Asunto(s)
Dolor Crónico/genética , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Dolor Crónico/metabolismo , Humanos
11.
Mol Pain ; 11: 32, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26024835

RESUMEN

Chronic pain is a major public health problem with limited treatment options. Opioids remain a routine treatment for chronic pain, but extended exposure to opioid therapy can produce opioid tolerance and hyperalgesia. Although the mechanisms underlying chronic pain, opioid-induced tolerance, and opioid-induced hyperalgesia remain to be uncovered, mammalian target of rapamycin (mTOR) is involved in these disorders. The mTOR complex 1 and its triggered protein translation are required for the initiation and maintenance of chronic pain (including cancer pain) and opioid-induced tolerance/hyperalgesia. Given that mTOR inhibitors are FDA-approved drugs and an mTOR inhibitor is approved for the treatment of several cancers, these findings suggest that mTOR inhibitors will likely have multiple clinical benefits, including anticancer, antinociception/anti-cancer pain, and antitolerance/hyperalgesia. This paper compares the role of mTOR complex 1 in chronic pain, opioid-induced tolerance, and opioid-induced hyperalgesia.


Asunto(s)
Analgésicos Opioides/metabolismo , Dolor Crónico/metabolismo , Tolerancia a Medicamentos/fisiología , Hiperalgesia/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos , Hiperalgesia/tratamiento farmacológico , Umbral del Dolor/fisiología
12.
Pain ; 156(4): 711-721, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25630025

RESUMEN

Peripheral nerve injury-induced changes in gene transcription and translation in primary sensory neurons of the dorsal root ganglion (DRG) are considered to contribute to neuropathic pain genesis. Transcription factors control gene expression. Peripheral nerve injury increases the expression of myeloid zinc finger protein 1 (MZF1), a transcription factor, and promotes its binding to the voltage-gated potassium 1.2 (Kv1.2) antisense (AS) RNA gene in the injured DRG. However, whether DRG MZF1 participates in neuropathic pain is still unknown. Here, we report that blocking the nerve injury-induced increase of DRG MZF1 through microinjection of MZF1 siRNA into the injured DRG attenuated the initiation and maintenance of mechanical, cold, and thermal pain hypersensitivities in rats with chronic constriction injury (CCI) of the sciatic nerve, without affecting locomotor functions and basal responses to acute mechanical, heat, and cold stimuli. Mimicking the nerve injury-induced increase of DRG MZF1 through microinjection of recombinant adeno-associated virus 5 expressing full-length MZF1 into the DRG produced significant mechanical, cold, and thermal pain hypersensitivities in naive rats. Mechanistically, MZF1 participated in CCI-induced reductions in Kv1.2 mRNA and protein and total Kv current and the CCI-induced increase in neuronal excitability through MZF1-triggered Kv1.2 AS RNA expression in the injured DRG neurons. MZF1 is likely an endogenous trigger of neuropathic pain and might serve as a potential target for preventing and treating this disorder.


Asunto(s)
Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/fisiología , Neuralgia/etiología , Neuralgia/patología , Traumatismos de los Nervios Periféricos/complicaciones , Transactivadores/metabolismo , Animales , ADN sin Sentido/farmacología , Ganglios Espinales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv.1.2/metabolismo , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Estimulación Física , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transactivadores/genética , Transducción Genética
13.
Anesthesiology ; 121(2): 409-17, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24739997

RESUMEN

Chronic pain, a common clinical symptom, is often treated inadequately or ineffectively in part due to the incomplete understanding of molecular mechanisms that initiate and maintain this disorder. Newly identified noncoding RNAs govern gene expression. Recent studies have shown that peripheral noxious stimuli drive expressional changes in noncoding RNAs and that these changes are associated with pain hypersensitivity under chronic pain conditions. This review first presents current evidence for the peripheral inflammation/nerve injury-induced change in the expression of two types of noncoding RNAs, microRNAs, and Kcna2 antisense RNA, in pain-related regions, particularly in the dorsal root ganglion. The authors then discuss how peripheral noxious stimuli induce such changes. The authors finally explore potential mechanisms of how expressional changes in dorsal root ganglion microRNAs and Kcna2 antisense RNA contribute to the development and maintenance of chronic pain. An understanding of these mechanisms may propose novel therapeutic strategies for preventing and/or treating chronic pain.


Asunto(s)
Dolor Crónico/genética , Dolor Crónico/fisiopatología , ARN no Traducido/genética , ARN no Traducido/fisiología , Humanos , Inflamación/genética , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Canal de Potasio Kv.1.2/biosíntesis , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv.1.2/fisiología , MicroARNs/biosíntesis , MicroARNs/genética , Neuralgia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...