Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Neuroinflammation ; 19(1): 144, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690777

RESUMEN

BACKGROUND: The adaption of brain region is fundamental to the development and maintenance of nervous system disorders. The prelimbic cortex (PrL) participates in the affective components of the pain sensation. However, whether and how the adaptation of PrL contributes to the comorbidity of neuropathic pain and depression are unknown. METHODS: Using resting-state functional magnetic resonance imaging (rs-fMRI), genetic knockdown or overexpression, we systematically investigated the activity of PrL region in the pathogenesis of neuropathic pain/depression comorbid using the combined approaches of immunohistochemistry, electrophysiology, and behavior. RESULTS: The activity of PrL and the excitability of pyramidal neurons were decreased, and the osteoclastic tartrate-resistant acid phosphatase 5 (Acp5) expression in PrL neurons was upregulated following the acquisition of spared nerve injury (SNI)-induced comorbidity. Genetic knockdown of Acp5 in pyramidal neurons, but not parvalbumin (PV) neurons or somatostatin (SST) neurons, attenuated the decrease of spike number, depression-like behavior and mechanical allodynia in comorbidity rats. Overexpression of Acp5 in PrL pyramidal neurons decreased the spike number and induced the comorbid-like behavior in naïve rats. Moreover, the expression of interleukin-6 (IL-6), phosphorylated STAT3 (p-STAT3) and acetylated histone H3 (Ac-H3) were significantly increased following the acquisition of comorbidity in rats. Increased binding of STAT3 to the Acp5 gene promoter and the interaction between STAT3 and p300 enhanced acetylation of histone H3 and facilitated the transcription of Acp5 in PrL in the modeled rodents. Inhibition of IL-6/STAT3 pathway prevented the Acp5 upregulation and attenuated the comorbid-like behaviors in rats. CONCLUSIONS: These data suggest that the adaptation of PrL mediated by IL-6/STAT3/Acp5 pathway contributed to the comorbidity of neuropathic pain/depression induced by SNI.


Asunto(s)
Interleucina-6 , Neuralgia , Fosfatasa Ácida/metabolismo , Animales , Comorbilidad , Depresión/metabolismo , Histonas , Interleucina-6/metabolismo , Neuralgia/metabolismo , Ratas , Factor de Transcripción STAT3/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo
2.
Neurochem Res ; 46(8): 2181-2191, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34032956

RESUMEN

Application of chemotherapeutic oxaliplatin represses gene transcription through induction of DNA methylation, which may contribute to oxaliplatin-induced chronic pain. Here, Ddr1, which showed an increased methylation in the promoter, was screened from the SRA methylation database (PRJNA587622) after oxaliplatin treatment. qPCR and MeDIP assays verified that oxaliplatin treatment increased the methylation in Ddr1 promoter region and decreased the expression of DDR1 in the neurons of spinal dorsal horn. In addition, overexpression of DDR1 by intraspinal injection of AAV-hSyn-Ddr1 significantly alleviated the mechanical allodynia induced by oxaliplatin. Furthermore, we found that oxaliplatin treatment increased the expression of DNMT3b and ZEB1 in dorsal horn neurons, and promoted the interaction between DNMT3b and ZEB1. Intrathecal injection of ZEB1 siRNA inhibited the enhanced recruitment of DNMT3b and the hypermethylation in Ddr1 promoter induced by oxaliplatin. Finally, ZEB1 siRNA rescued the DDR1 downregulation and mechanical allodynia induced by oxaliplatin. In conclusion, these results suggested that the ZEB1 recruited DNMT3b to the Ddr1 promoter, which induced the DDR1 downregulation and contributed to the oxaliplatin-induced chronic pain.


Asunto(s)
Dolor Crónico/metabolismo , Metilación de ADN/fisiología , Receptor con Dominio Discoidina 1/genética , Oxaliplatino/efectos adversos , Asta Dorsal de la Médula Espinal/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Dolor Crónico/inducido químicamente , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Regiones Promotoras Genéticas/fisiología , ARN Interferente Pequeño/farmacología , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , ADN Metiltransferasa 3B
3.
Brain Behav Immun ; 84: 72-79, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31751616

RESUMEN

Conditioned place preference (CPP) is a learned behavior, in which animals learn to associate environmental contexts with rewarding effects. The formation of CPP is an integrated outcome of multiple learning processes. Although multiple anatomical substrates underlying this contextual learning have been proposed, it remains unknown whether a specific molecular signaling pathway within CA1 mediates context learning associated with morphine conditioning. Here, we showed that repeated context learning associated with morphine conditioning significantly increased CXCL12 levels in hippocampal CA1 neurons, and the inhibition of CXCL12 expression ameliorated the CPP behavior following context exposure with morphine conditioning. Additionally, repeated context exposure with morphine conditioning increased the phosphorylation of STAT3 and the acetylation of histone H4 in CXCL12-expressing neurons in CA1. Immunoprecipitation and chromatin immunoprecipitation assays demonstrated that repeated context exposure with morphine conditioning increased the binding of STAT3 to the CXCL12 gene promoter and the interaction between STAT3 and p300, which contributed to the enhanced transcription of CXCL12 by increasing the acetylation of histone H4 in the CXCL12 gene promoter. The inhibition of STAT3 by intrathecal injection of S3I-201 suppressed the acetylation of histone H4. These data demonstrated the epigenetic upregulation of CXCL12 following repeated context exposure with morphine conditioning, which potentially contributed to the spatial memory consolidation associated with conditioned place preference induced by morphine conditioning.


Asunto(s)
Quimiocina CXCL12/genética , Condicionamiento Psicológico , Epigénesis Genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Morfina/farmacología , Memoria Espacial/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/metabolismo , Masculino , Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley
4.
J Neuroinflammation ; 16(1): 29, 2019 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-30736806

RESUMEN

BACKGROUND: Studies showed that upregulation of Nav1.6 increased the neuronal excitability and participated in neuropathic pain in the dorsal root ganglion (DRG). However, the molecular mechanisms underlying Nav1.6 upregulation were not reported yet. METHODS: The paw withdrawal threshold was measured in the rodents following lumbar 5 ventral root transection (L5-VRT). Then qPCR, western blotting, immunoprecipitation, immunohistochemistry, and chromatin immunoprecipitation assays were performed to explore the molecular mechanisms in vivo and in vitro. RESULTS: We found that the levels of Nav1.6 and phosphorylated STAT3 were significantly increased in DRG neurons following L5-VRT, and TNF-α incubation also upregulated the Nav1.6 expression in cultured DRG neurons. Furthermore, immunoprecipitation and chromatin immunoprecipitation assays demonstrated that L5-VRT increased the binding of STAT3 to the Scn8a (encoding Nav1.6) promoter and the interaction between STAT3 and p300, which contributed to the enhanced transcription of Scn8a by increasing histone H4 acetylation in Scn8a promoter in DRG. Importantly, intraperitoneal injection of the TNF-α inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. CONCLUSION: These results suggested a new mechanism for Nav1.6 upregulation involving TNF-α/STAT3 pathway activation and subsequent STAT3-mediated histone H4 hyperacetylation in the Scn8a promoter region in DRG, which contributed to L5-VRT-induced neuropathic pain.


Asunto(s)
Epigénesis Genética/genética , Ganglios Espinales/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/biosíntesis , Neuralgia/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/fisiopatología , Inmunohistoquímica , Masculino , Neuralgia/fisiopatología , Ratas , Ratas Sprague-Dawley , Raíces Nerviosas Espinales
5.
J Neurosci ; 36(23): 6321-31, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27277808

RESUMEN

UNLABELLED: Neuropathic pain is a common neurobiological disease involving multifaceted maladaptations ranging from gene modulation to synaptic dysfunction, but the interactions between synaptic dysfunction and the genes that are involved in persistent pain remain elusive. In the present study, we found that neuropathic pain induced by the chemotherapeutic drug paclitaxel or L5 ventral root transection significantly impaired the function of GABAergic synapses of spinal dorsal horn neurons via the reduction of the GAD67 expression. We also found that mir-500 expression was significantly increased and involved in the modulation of GAD67 expression via targeting the specific site of Gad1 gene in the dorsal horn. In addition, knock-out of mir-500 or using mir-500 antagomir rescued the GABAergic synapses in the spinal dorsal horn neurons and attenuated the sensitized pain behavior in the rats with neuropathic pain. To our knowledge, this is the first study to investigate the function significance and the underlying molecular mechanisms of mir-500 in the process of neuropathic pain, which sheds light on the development of novel therapeutic options for neuropathic pain. SIGNIFICANCE STATEMENT: Neuropathic pain is a common neurobiological disease involving multifaceted maladaptations ranging from gene modulation to synaptic dysfunction, but the underlying molecular mechanisms remain elusive. The present study illustrates for the first time a mir-500-mediated mechanism underlying spinal GABAergic dysfunction and sensitized pain behavior in neuropathic pain induced by the chemotherapeutic drug paclitaxel or L5 ventral root transection, which sheds light on the development of novel therapeutic options for neuropathic pain.


Asunto(s)
Regulación hacia Abajo/genética , Glutamato Descarboxilasa/metabolismo , MicroARNs/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Antagomirs/farmacología , Antineoplásicos Fitogénicos/farmacología , Modelos Animales de Enfermedad , Glutamato Descarboxilasa/genética , Hiperalgesia , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neuralgia/etiología , Paclitaxel/farmacología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo
6.
Anesthesiology ; 124(5): 1109-21, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26978408

RESUMEN

BACKGROUND: Systemic administration of oxaliplatin has no effect on the tumors in the central nervous system (CNS) due to the limited concentration of oxaliplatin in the cerebrospinal fluid (CSF), while it was clinically reported that oxaliplatin can induce acute encephalopathy. Currently, the impairment of neuronal functions in the CNS after systemic administration of oxaliplatin remains uninvestigated. METHODS: The von Frey test and the plantar test were performed to evaluate neuropathic pain behavior after a single intraperitoneal administration of oxaliplatin (4 mg/kg) in rats. Inductively coupled plasma-mass spectrometry, electrophysiologic recording, real-time quantitative reverse transcription polymerase chain reaction, chromatin immunoprecipitation, Western blot, immunohistochemistry, and small interfering RNA were applied to understand the mechanisms. RESULTS: Concentration of oxaliplatin in CSF showed a time-dependent increase after a single administration of oxaliplatin. Spinal application of oxaliplatin at the detected concentration (6.6 nM) significantly increased the field potentials in the dorsal horn, induced acute mechanical allodynia (n = 12 each) and thermal hyperalgesia (n = 12 each), and enhanced the evoked excitatory postsynaptic currents and spontaneous excitatory postsynaptic currents in the projection neurokinin 1 receptor-expressing lamina I to II neurons. The authors further found that oxaliplatin significantly increased the nuclear factor-κB p65 binding and histone H4 acetylation in cx3cl1 promoter region. Thus, the upregulated spinal CX3CL1 markedly mediated the induction of central sensitization and acute pain behavior after oxaliplatin administration. CONCLUSIONS: The findings of this study suggested that oxaliplatin in CSF may directly impair the normal function of central neurons and contribute to the rapid development of CNS-related side effects during chemotherapy. This provides novel targets to prevent oxaliplatin-induced acute painful neuropathy and encephalopathy.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/líquido cefalorraquídeo , Neuralgia/inducido químicamente , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/líquido cefalorraquídeo , Animales , Quimiocina CX3CL1/metabolismo , Histonas/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Inyecciones Intraperitoneales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxaliplatino , Dimensión del Dolor , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo , Factor de Transcripción ReIA/metabolismo
7.
Anesthesiology ; 122(5): 1142-51, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25494456

RESUMEN

BACKGROUND: Up-regulation of CX3CL1 has been revealed to be involved in the neuropathic pain induced by nerve injury. However, whether CX3CL1 participates in the paclitaxel-induced painful peripheral neuropathy remains unknown. The aim of the current study was to elucidate the involvement of transcriptional factors nuclear factor-κB (NF-κB) and its causal interaction with CX3CL1 signaling in the paclitaxel-induced painful peripheral neuropathy. METHODS: Painful peripheral neuropathy induced by paclitaxel treatment was established in adult male Sprague-Dawley rats. The von Frey test were performed to evaluate neuropathic pain behavior, and real-time quantitative reverse transcription polymerase chain reaction, chromatin immunoprecipitation, Western blot, immunohistochemistry, and small interfering RNA were performed to understand the molecular mechanisms. RESULTS: The application of paclitaxel induced an up-regulation of CX3CL1 expression in the spinal neurons, which is reduced significantly by NF-κB inhibitor ammonium pyrrolidinedithiocarbamate or p65 small interfering RNA. Blockade of either CX3CL1 (n = 12 each) or NF-κB (n = 12 each) signaling pathway attenuated mechanical allodynia induced by paclitaxel. Chromatin immunoprecipitation further found that paclitaxel induced an increased recruitment of nuclear factor-κB (NF-κB)p65 to the Cx3cl1 promoter region. Furthermore, an increased acetylation level of H4, but not H3, in Cx3cl1 promoter region in spinal neurons was detected after paclitaxel treatment, which was reversed by inhibition of NF-κB with ammonium pyrrolidinedithiocarbamate or p65 small interfering RNA. CONCLUSIONS: These findings suggest that up-regulation of CX3CL1 via NF-κB-dependent H4 acetylation might be critical for paclitaxel-induced mechanical allodynia.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Quimiocina CX3CL1/biosíntesis , Histonas/metabolismo , FN-kappa B/biosíntesis , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/genética , Acetilación , Animales , Quimiocina CX3CL1/genética , Citocinas/biosíntesis , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Masculino , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Dimensión del Dolor/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/metabolismo , Pirrolidinas/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Tiocarbamatos/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/biosíntesis , Factor de Transcripción ReIA/genética
8.
Brain Behav Immun ; 38: 185-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24530998

RESUMEN

Bortezomib (BTZ) is a frequently used chemotherapeutic drug for the treatment of refractory multiple myeloma and hematological neoplasms. The mechanism by which the administration of BTZ leads to painful peripheral neuropathy remains unclear. In the present study, we first determined that the administration of BTZ upregulated the expression of TNF-α and phosphorylated JNK1/2 in the dorsal root ganglion (DRG) of rat. Furthermore, the TNF-α synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Knockout of the expression of TNF-α receptor TNFR1 (TNFR1 KO mice) or TNFR2 (TNFR2 KO mice) inhibited JNK1 and JNK2 activation and decreased mechanical allodynia induced by BTZ. These results suggest that upregulated TNF-α expression may activate JNK signaling via TNFR1 or TNFR2 to mediate mechanical allodynia following BTZ treatment.


Asunto(s)
Antineoplásicos/toxicidad , Ácidos Borónicos/toxicidad , Ganglios Espinales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Pirazinas/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Bortezomib , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/enzimología , Enfermedades del Sistema Nervioso Periférico/enzimología , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Front Neurosci ; 18: 1428666, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39308951

RESUMEN

Introduction: It is well known that there are significant differences in the prevalence of chronic pain between males and females. Human and animal imaging studies have shown that chronic pain profoundly alters the structure and function of brain regions. However, there is limited research on the sex-specific mechanisms underlying the brain plasticity and adaptive changes associated with chronic pain. In this article, we conducted a multimodal study to evaluate how nerve injury-induced chronic pain affects the brain. Methods: Male and female Sprague-Dawley (SD) rats with spared nerve injury (SNI) model underwent resting-state functional magnetic resonance imaging (rs-fMRI) (male sham group: n = 18; male SNI group: n = 18; female sham group: n = 20; female SNI group: n = 18) and magnetic resonance diffusion tensor imaging (DTI) (male sham group: n = 23; male SNI group: n = 21; female sham group: n = 20; female SNI group: n = 21) scanning. ICA method, Fractional amplitude of low-frequency fluctuations (fALFF), immunofluorescence staining, and graph theory analysis was utilized to extract the rs-fMRI changes of brain regions of each group. Results: Using SNI model, which promotes long-lasting mechanical allodynia, we found that neuropathic pain deeply modified the intrinsic organization of the brain functional network in male and female rats (main effect of operation: F = 298.449, P < 0.001). 64 independent components (ICs) in the brain were divided and assigned to 16 systems. In male rats, we observed significant alterations in the microstructure of the hippocampal cornu ammonis 1 and cornu ammonis 2 (CA1/CA2) region, as indicated by increased mean diffusivity (MD) (CA1_L: P = 0.02; CA1_R: P = 0.031; CA2_L: P = 0.035; CA2_R: P = 0.015) and radial diffusivity (RD) (CA1_L: P = 0.028; CA1_R: P = 0.033; CA2_L: P = 0.037; CA2_R: P = 0.038) values, along with enhanced activating transcription factor 3 (ATF3) expression. Conversely, in female rats, we found significant increases in the fractional amplitude of low frequency fluctuations (fALFF) value within the hippocampal dentate gyrus (DG) (F = 5.419, P = 0.023), accompanied by elevated c-Fos signal (F = 6.269, P = 0.031). Furthermore, graph theory analysis revealed notable differences in the small-world network of the hippocampal system in female rats, characterized by reduced small-world attributes and increased inter-nodal transmission efficiency. Discussion: Our study indicates sex differences in structural and functional alterations in the hippocampal system in rats under chronic pain conditions. The results suggest that the hippocampus system plays an important role in the different mechanisms of chronic pain in different sexes. These findings provide reliable insights to explore the complex mechanisms underlying sex differences in chronic pain.

10.
Adv Sci (Weinh) ; 11(34): e2401855, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38973158

RESUMEN

Clinically, chronic pain and depression often coexist in multiple diseases and reciprocally reinforce each other, which greatly escalates the difficulty of treatment. The neural circuit mechanism underlying the chronic pain/depression comorbidity remains unclear. The present study reports that two distinct subregions in the paraventricular thalamus (PVT) play different roles in this pathological process. In the first subregion PVT posterior (PVP), glutamatergic neurons (PVPGlu) send signals to GABAergic neurons (VLPAGGABA) in the ventrolateral periaqueductal gray (VLPAG), which mediates painful behavior in comorbidity. Meanwhile, in another subregion PVT anterior (PVA), glutamatergic neurons (PVAGlu) send signals to the nucleus accumbens D1-positive neurons and D2-positive neurons (NAcD1→D2), which is involved in depression-like behavior in comorbidity. This study demonstrates that the distinct thalamo-subcortical circuits PVPGlu→VLPAGGABA and PVAGlu→NAcD1→D2 mediated painful behavior and depression-like behavior following spared nerve injury (SNI), respectively, which provides the circuit-based potential targets for preventing and treating comorbidity.


Asunto(s)
Conducta Animal , Depresión , Modelos Animales de Enfermedad , Tálamo , Animales , Depresión/fisiopatología , Masculino , Tálamo/fisiopatología , Conducta Animal/fisiología , Ratones , Vías Nerviosas/fisiopatología , Dolor/fisiopatología , Dolor Crónico/fisiopatología
11.
Neurosci Bull ; 39(6): 947-961, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36637791

RESUMEN

Effective treatments for neuropathic pain are lacking due to our limited understanding of the mechanisms. The circRNAs are mainly enriched in the central nervous system. However, their function in various physiological and pathological conditions have yet to be determined. Here, we identified circFhit, an exon-intron circRNA expressed in GABAergic neurons, which reduced the inhibitory synaptic transmission in the spinal dorsal horn to mediate spared nerve injury-induced neuropathic pain. Moreover, we found that circFhit decreased the expression of GAD65 and induced hyperexcitation in NK1R+ neurons by promoting the expression of its parental gene Fhit in cis. Mechanistically, circFhit was directly bound to the intronic region of Fhit, and formed a circFhit/HNRNPK complex to promote Pol II phosphorylation and H2B monoubiquitination by recruiting CDK9 and RNF40 to the Fhit intron. In summary, we revealed that the exon-intron circFhit contributes to GABAergic neuron-mediated NK1R+ neuronal hyperexcitation and neuropathic pain via regulating Fhit in cis.


Asunto(s)
Neuralgia , Células del Asta Posterior , Ratas , Animales , Células del Asta Posterior/metabolismo , Células del Asta Posterior/patología , Asta Dorsal de la Médula Espinal/metabolismo , Transmisión Sináptica
12.
Sci Adv ; 9(25): eadg5849, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37352353

RESUMEN

The association between rewarding and drug-related memory is a leading factor for the formation of addiction, yet the neural circuits underlying the association remain unclear. Here, we showed that the interstitial nucleus of the posterior limb of the anterior commissure (IPAC) integrated rewarding and environmental memory information by two different receiving projections from ventral tegmental area (VTA) and nucleus accumbens shell region (NAcSh) to mediate the acquisition of morphine conditioned place preference (CPP). A projection from the VTA GABAergic neurons (VTAGABA) to the IPAC lateral region GABAergic neurons (IPACLGABA) mediated the effect of morphine rewarding, whereas the pathway from NAcSh dopamine receptor 1-expressing neurons (NAcShD1) to the IPAC medial region GABAergic neurons (IPACMGABA) was involved in the acquisition of environmental memory. These findings demonstrated that the distinct IPAC circuits VTAGABA→IPACLGABA and NAcShD1R→IPACMGABA were attributable to the rewarding and environmental memory during the acquisition of morphine CPP, respectively, and provided the circuit-based potential targets for preventing and treating opioid addiction.


Asunto(s)
Morfina , Área Tegmental Ventral , Morfina/farmacología , Recompensa , Neuronas GABAérgicas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
13.
Sci Signal ; 11(523)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588412

RESUMEN

Palmitoylation of δ-catenin is critical to synapse plasticity and memory formation. We found that δ-catenin palmitoylation is also instrumental in the development of neuropathic pain. The abundances of palmitoylated δ-catenin and the palmitoyl acyltransferase DHHC3 were increased in dorsal root ganglion (DRG) sensory neurons in rat models of neuropathic pain. Inhibiting palmitoyl acyltransferases or decreasing δ-catenin abundance in the DRG by intrathecal injection of 2-bromopalmitate or shRNA, respectively, alleviated oxaliplatin or nerve injury-induced neuropathic pain in the rats. The palmitoylation of δ-catenin, which was induced by the inflammatory cytokine TNF-α, facilitated its interaction with the voltage-gated sodium channel Nav1.6 and the kinesin motor protein KIF3A, which promoted the trafficking of Nav1.6 to the plasma membrane in DRG neurons and contributed to mechanical hypersensitivity and allodynia in rats. These findings suggest that a palmitoylation-mediated KIF3A/δ-catenin/Nav1.6 complex enhances the transmission of mechanical and nociceptive signals; thus, blocking this mechanism may be therapeutic in patients with neuropathic pain.


Asunto(s)
Cateninas/metabolismo , Membrana Celular/metabolismo , Cinesinas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuralgia/fisiopatología , Células Receptoras Sensoriales/fisiología , Animales , Ganglios Espinales/citología , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Lipoilación , Masculino , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Oxaliplatino , Palmitatos/administración & dosificación , Palmitatos/farmacología , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Catenina delta
14.
Exp Neurol ; 296: 74-82, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28729113

RESUMEN

Bortezomib is a first-line chemotherapeutic drug widely used for multiple myeloma and other nonsolid malignancies. Although bortezomib-induced persistent pain is easily diagnosed in clinic, the pathogenic mechanism remains unclear. Here, we studied this issue with use of a rat model of systemic intraperitoneal administration of bortezomib for consecutive 5days. Consisted with our previous study, we found that bortezomib treatment markedly induced mechanical allodynia in rats. Furthermore, we first found that bortezomib treatment significantly induced the upregulation of methylglyoxal in spinal dorsal horn of rats. Spinal local application of methylglyoxal also induced mechanical allodynia and central sensitization in normal rats. Moreover, administration of bortezomib upregulated the expression of receptors for advanced glycation end products (RAGE) and phosphorylated STAT3 (p-STAT3) in dorsal horn. Importantly, intrathecal injection of metformin, a known scavenger of methylglyoxal, significantly attenuated the upregulation of methylglyoxal and RAGE in dorsal horn, central sensitization and mechanical allodynia induced by bortezomib treatment, and blockage of RAGE also prevented the upregulation of p-STAT3, central sensitization and mechanical allodynia induced by bortezomib treatment. In addition, inhibition of STAT3 activity by S3I-201 attenuated bortezomib-induced mechanical allodynia and central sensitization. Local knockdown of STAT3 also ameliorated the mechanical allodynia induced by bortezomib administration. Our results suggest that accumulation of methylglyoxal may activate the RAGE/STAT3 signaling pathway in dorsal horn, and contributes to the spinal central sensitization and persistent pain induced by bortezomib treatment.


Asunto(s)
Bortezomib/toxicidad , Sensibilización del Sistema Nervioso Central/efectos de los fármacos , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Piruvaldehído/farmacología , Piruvaldehído/uso terapéutico , Médula Espinal/fisiopatología , Animales , Antineoplásicos/toxicidad , Modelos Animales de Enfermedad , Masculino , Fibras Nerviosas Amielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/fisiología , Dolor/patología , Dimensión del Dolor/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética , Transducción Genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
15.
Pain ; 158(4): 637-648, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28072604

RESUMEN

Clinically, Microtubule-targeted agents-induced neuropathic pain hampers chemotherapeutics for patients with cancer. Here, we found that application of paclitaxel or vincristine increased the protein and mRNA expression of CXCL12 and frequency and amplitude of miniature excitatory post synaptic currents (mEPSCs) in spinal dorsal horn neurons. Spinal local application of CXCL12 induced the long-term potentiation of nociceptive synaptic transmission and increased the amplitude of mEPSCs. Inhibition of CXCL12 using the transgenic mice (CXCL12) or neutralizing antibody or siRNA ameliorated the mEPSC's enhancement and mechanical allodynia. In addition, paclitaxel and vincristine both could increase the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the acetylation of histone H4 in the CXCL12-expressing neurons. Immunoprecipitation and chromatin immunoprecipitation assays demonstrated that antitubulin chemotherapeutics increased the binding of STAT3 to the CXCL12 gene promoter and the interaction between STAT3 and p300, and contributed to the enhanced transcription of CXCL12 by increasing the acetylation of histone H4 in CXCL12 gene promoter. Inhibition of STAT3 by intrathecal injection of adeno-associated virus encoding Cre and green fluorescent protein into STAT3 mice or inhibitor S3I-201 into rats suppressed the CXCL12 upsurge by decreasing the acetylation of histone H4. Finally, blockade of CXCR4 but not CXCR7 ameliorated the paclitaxel- or vincristine-induced mechanical allodynia. Together, these results suggested that enhanced interaction between STAT3 and p300 mediated the epigenetic upregulation of CXCL12 in dorsal horn neurons, which contributed to the antitubulin chemotherapeutics-induced persistent pain.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Tubulina (Proteína)/inmunología , Regulación hacia Arriba/efectos de los fármacos , Animales , Anticuerpos/uso terapéutico , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Modelos Animales de Enfermedad , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/genética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Paclitaxel , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Factores de Tiempo , Regulación hacia Arriba/genética , Vincristina
16.
Exp Neurol ; 288: 85-93, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27856287

RESUMEN

Our recent findings demonstrated that oxaliplatin entering CNS may directly induce spinal central sensitization, and contribute to the rapid development of CNS-related side effects including acute pain during chemotherapy. However, the mechanism is largely unclear. In the current study, we found that the amplitude of C-fiber-evoked field potentials was significantly increased and the expression of phosphorylated mammalian AMP-activated protein kinase α (AMPKα) was markedly decreased following high frequency stimulation (HFS) or single intraperitoneal injection of oxaliplatin (4mg/kg). Spinal local application of AMPK agonist metformin (25µg) prevented the long term potentiation (LTP) induction and the activation of mTOR/p70S6K signal pathway, and significantly attenuated the acute thermal hyperalgesia and mechanical allodynia following single oxaliplatin treatment. Importantly, we found that incubation of low concentration oxaliplatin at dose of 6.6nM (the detected concentration in CSF following a single intraperitoneal injection of oxaliplatin) also significantly inhibited the AMPKα activation and increased the amplitude of sEPSCs, the number of action potential, and the expression of p-mTOR and p-p70S6K in spinal cord slices. Metformin (25µg) or rapamycin (2µg) inhibited the increased excitability of dorsal horn neurons and the decrease of p-AMPKα expression induced by low concentration oxaliplatin incubation. Furthermore, spinal application of AMPK inhibitor compound C (5µg) induced the spinal LTP, thermal hyperalgesia and mechanical allodynia, and rapamycin attenuated the spinal LTP, the thermal hyperalgesia and mechanical allodynia following oxaliplatin treatment (i.p.). Local application of metformin significantly decreased the mTOR and p70S6K activation induced by tetanus stimulation or oxaliplatin (i.p.). These results suggested that the decreased AMPKα activity via negatively regulating mTOR/p70S6K signal pathway enhanced the synaptic plasticity and contributed to acute pain induced by low concentration of oxaliplatin entering CNS.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dolor Agudo , Plasticidad Neuronal/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Transducción de Señal/efectos de los fármacos , Dolor Agudo/inducido químicamente , Dolor Agudo/patología , Dolor Agudo/fisiopatología , Animales , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Potenciales Evocados/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/fisiopatología , Técnicas In Vitro , Masculino , Fibras Nerviosas Amielínicas/efectos de los fármacos , Oxaliplatino , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Médula Espinal/patología , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
17.
Neural Regen Res ; 11(12): 1969-1975, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28197194

RESUMEN

Striatal neurons can be either projection neurons or interneurons, with each type exhibiting distinct susceptibility to various types of brain damage. In this study, 6-hydroxydopamine was injected into the right medial forebrain bundle to induce dopamine depletion, and/or ibotenic acid was injected into the M1 cortex to induce motor cortex lesions. Immunohistochemistry and western blot assay showed that dopaminergic depletion results in significant loss of striatal projection neurons marked by dopamine- and cyclic adenosine monophosphate-regulated phosphoprotein, molecular weight 32 kDa, calbindin, and µ-opioid receptor, while cortical lesions reversed these pathological changes. After dopaminergic deletion, the number of neuropeptide Y-positive striatal interneurons markedly increased, which was also inhibited by cortical lesioning. No noticeable change in the number of parvalbumin-positive interneurons was found in 6-hydroxydopamine-treated rats. Striatal projection neurons and interneurons show different susceptibility to dopaminergic depletion. Further, cortical lesions inhibit striatal dysfunction and damage induced by 6-hydroxydopamine, which provides a new possibility for clinical treatment of Parkinson's disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA