Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 768
Filtrar
1.
ACS Chem Biol ; 19(7): 1626-1637, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39026469

RESUMEN

Chronic pain is a prevalent problem that plagues modern society, and better understanding its mechanisms is critical for developing effective therapeutics. Nerve growth factor (NGF) and its primary receptor, Tropomyosin receptor kinase A (TrkA), are known to be potent mediators of chronic pain, but there is a lack of established methods for precisely perturbing the NGF/TrkA signaling pathway in the study of pain and nociception. Optobiological tools that leverage light-induced protein-protein interactions allow for precise spatial and temporal control of receptor signaling. Previously, our lab reported a blue light-activated version of TrkA generated using light-induced dimerization of the intracellular TrkA domain, opto-iTrkA. In this work, we show that opto-iTrkA activation is able to activate endogenous ERK and Akt signaling pathways and causes the retrograde transduction of phospho-ERK signals in dorsal root ganglion (DRG) neurons. Opto-iTrkA activation also sensitizes the transient receptor potential vanilloid 1 (TRPV1) channel in cellular models, further corroborating the physiological relevance of the optobiological stimulus. Finally, we show that opto-iTrkA enables light-inducible potentiation of mechanical sensitization in mice. Light illumination enables nontraumatic and reversible (<2 days) sensitization of mechanical pain in mice transduced with opto-iTrkA, which provides a platform for dissecting TrkA pathways for nociception in vitro and in vivo.


Asunto(s)
Dolor Crónico , Ganglios Espinales , Luz , Receptor trkA , Animales , Receptor trkA/metabolismo , Dolor Crónico/metabolismo , Ratones , Ganglios Espinales/metabolismo , Canales Catiónicos TRPV/metabolismo , Humanos , Transducción de Señal , Ratones Endogámicos C57BL , Masculino , Factor de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo
2.
CNS Neurosci Ther ; 30(7): e14831, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38961317

RESUMEN

AIMS: Comorbid anxiodepressive-like symptoms (CADS) in chronic pain are closely related to the overactivation of the lateral habenula (LHb). Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have been implicated to play a key role in regulating neuronal excitability. However, the role of HCN channels in the LHb during CADS has not yet been characterized. This study aimed to investigate the effect of HCN channels in the LHb on CADS during chronic pain. METHODS: After chronic neuropathic pain induction by spared nerve injury (SNI), mice underwent a sucrose preference test, forced swimming test, tail suspension test, open-field test, and elevated plus maze test to evaluate their anxiodepressive-like behaviors. Electrophysiological recordings, immunohistochemistry, Western blotting, pharmacological experiments, and virus knockdown strategies were used to investigate the underlying mechanisms. RESULTS: Evident anxiodepressive-like behaviors were observed 6w after the SNI surgery, accompanied by increased neuronal excitability, enhanced HCN channel function, and increased expression of HCN2 isoforms in the LHb. Either pharmacological inhibition or virus knockdown of HCN2 channels significantly reduced LHb neuronal excitability and ameliorated both pain and depressive-like behaviors. CONCLUSION: Our results indicated that the LHb neurons were hyperactive under CADS in chronic pain, and this hyperactivation possibly resulted from the enhanced function of HCN channels and up-regulation of HCN2 isoforms.


Asunto(s)
Depresión , Habénula , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Animales , Habénula/metabolismo , Habénula/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ratones , Masculino , Depresión/metabolismo , Neuralgia/metabolismo , Neuralgia/psicología , Ratones Endogámicos C57BL , Dolor Crónico/metabolismo , Dolor Crónico/psicología , Canales de Potasio
3.
Sci Signal ; 17(847): eadn8936, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39078919

RESUMEN

Obstructive sleep apnea (OSA) is a prevalent sleep disorder that is associated with increased incidence of chronic musculoskeletal pain. We investigated the mechanism of this association in a mouse model of chronic intermittent hypoxia (CIH) that mimics the repetitive hypoxemias of OSA. After 14 days of CIH, both male and female mice exhibited behaviors indicative of persistent pain, with biochemical markers in the spinal cord dorsal horn and sensory neurons of the dorsal root ganglia consistent with hyperalgesic priming. CIH, but not sleep fragmentation alone, induced an increase in macrophage recruitment to peripheral sensory tissues (sciatic nerve and dorsal root ganglia), an increase in inflammatory cytokines in the circulation, and nociceptor sensitization. Peripheral macrophage ablation blocked CIH-induced hyperalgesic priming. The findings suggest that correcting the hypoxia or targeting macrophage signaling might suppress persistent pain in patients with OSA.


Asunto(s)
Hipoxia , Macrófagos , Nociceptores , Animales , Hipoxia/metabolismo , Macrófagos/metabolismo , Masculino , Femenino , Ratones , Nociceptores/metabolismo , Ganglios Espinales/metabolismo , Apnea Obstructiva del Sueño/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Citocinas/metabolismo , Dolor Crónico/metabolismo , Dolor Crónico/inmunología
4.
Sci Rep ; 14(1): 14715, 2024 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926482

RESUMEN

Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90ß, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90ß or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90ß) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.


Asunto(s)
Analgésicos Opioides , Proteínas HSP90 de Choque Térmico , Morfina , Médula Espinal , Animales , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Ratones , Analgésicos Opioides/farmacología , Masculino , Femenino , Morfina/farmacología , Isoformas de Proteínas/metabolismo , Tolerancia a Medicamentos , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Modelos Animales de Enfermedad , Inyecciones Espinales
5.
Biomed Pharmacother ; 176: 116856, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38852510

RESUMEN

Fibromyalgia is characterised by widespread chronic pain and is often accompanied by comorbidities such as sleep disorders, anxiety, and depression. Because it is often accompanied by many adverse symptoms and lack of effective treatment, it is important to search for the pathogenesis and treatment of fibromyalgia. Astaxanthin, a carotenoid pigment known for its anti-inflammatory and antioxidant properties, has demonstrated effective analgesic effects in neuropathic pain. However, its impact on fibromyalgia remains unclear. Therefore, in this study, we constructed a mouse model of fibromyalgia and investigated the effect of astaxanthin on chronic pain and associated symptoms through multiple intragastrical injections. We conducted behavioural assessments to detect pain and depression-like states in mice, recorded electroencephalograms to monitor sleep stages, examined c-Fos activation in the anterior cingulate cortex, measured activation of spinal glial cells, and assessed levels of inflammatory factors in the brain and spinal cord, including interleukin (IL)-1ß, IL-6, and tumour necrosis factor- α(TNF-α).Additionally, we analysed the expression levels of IL-6, IL-10, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), Apoptosis-associated speck-like protein containing CARD, and Caspase-1 proteins. The findings revealed that astaxanthin significantly ameliorated mechanical and thermal pain in mice with fibromyalgia and mitigated sleep disorders and depressive-like symptoms induced by pain. A potential mechanism underlying these effects is the anti-inflammatory action of astaxanthin, likely mediated through the inhibition of the NLRP3 inflammasome, which could be one of the pathways through which astaxanthin alleviates fibromyalgia. In conclusion, our study suggests that astaxanthin holds promise as a potential analgesic medication for managing fibromyalgia and its associated symptoms.


Asunto(s)
Depresión , Fibromialgia , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Xantófilas , Animales , Xantófilas/farmacología , Fibromialgia/tratamiento farmacológico , Fibromialgia/complicaciones , Fibromialgia/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Inflamasomas/metabolismo , Inflamasomas/antagonistas & inhibidores , Depresión/tratamiento farmacológico , Depresión/metabolismo , Ratones , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Analgésicos/farmacología , Antiinflamatorios/farmacología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Citocinas/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Conducta Animal/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 121(27): e2403777121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38916998

RESUMEN

Spinal cord dorsal horn inhibition is critical to the processing of sensory inputs, and its impairment leads to mechanical allodynia. How this decreased inhibition occurs and whether its restoration alleviates allodynic pain are poorly understood. Here, we show that a critical step in the loss of inhibitory tone is the change in the firing pattern of inhibitory parvalbumin (PV)-expressing neurons (PVNs). Our results show that PV, a calcium-binding protein, controls the firing activity of PVNs by enabling them to sustain high-frequency tonic firing patterns. Upon nerve injury, PVNs transition to adaptive firing and decrease their PV expression. Interestingly, decreased PV is necessary and sufficient for the development of mechanical allodynia and the transition of PVNs to adaptive firing. This transition of the firing pattern is due to the recruitment of calcium-activated potassium (SK) channels, and blocking them during chronic pain restores normal tonic firing and alleviates chronic pain. Our findings indicate that PV is essential for controlling the firing pattern of PVNs and for preventing allodynia. Developing approaches to manipulate these mechanisms may lead to different strategies for chronic pain relief.


Asunto(s)
Dolor Crónico , Parvalbúminas , Parvalbúminas/metabolismo , Animales , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Potenciales de Acción/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
7.
Neuroscience ; 548: 39-49, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38697463

RESUMEN

Chronic inflammatory pain is the highest priority for people with osteoarthritis when seeking medical attention. Despite the availability of NSAIDs and glucocorticoids, central sensitization and peripheral sensitization make pain increasingly difficult to control. Previous studies have identified the ubiquitination system as an important role in the chronic inflammatory pain. Our study displayed that the E3 ubiquitin ligase tripartite motif-containing 14 (Trim14) was abnormally elevated in the serum of patients with osteoarthritis and pain, and the degree of pain was positively correlated with the degree of Trim14 elevation. Furthermore, CFA-induced inflammatory pain rat model showed that Trim14 was significantly increased in the L3-5 spinal dorsal horn (SDH) and dorsal root ganglion (DRG), and in turn the inhibitor of nuclear factor Kappa-B isoform α (IκBα) was decreased after Trim14 elevation. After intrathecal injection of Trim14 siRNA to inhibit Trim14 expression, IκBα expression was reversed and increased, and the pain behaviors and anxiety behaviors of rats were significantly relieved. Overall, these findings suggested that Trim14 may contribute to chronic inflammatory pain by degrading IκBα, and that Trim14 may become a novel therapeutic target for chronic inflammatory pain.


Asunto(s)
Dolor Crónico , Inflamación , Inhibidor NF-kappaB alfa , Osteoartritis , Ratas Sprague-Dawley , Transducción de Señal , Anciano , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ratas , Dolor Crónico/metabolismo , Ganglios Espinales/metabolismo , Inflamación/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Osteoartritis/metabolismo , Transducción de Señal/fisiología , Asta Dorsal de la Médula Espinal/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo
8.
Neuropharmacology ; 254: 109992, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38723742

RESUMEN

Chronic primary pain, characterized by overlapping symptoms of chronic pain, anxiety, and depression, is strongly associated with stress and is particularly prevalent among females. Recent research has convincingly linked epigenetic modifications in the medial prefrontal cortex (mPFC) to chronic pain and chronic stress. However, our understanding of the role of histone demethylation in the mPFC in chronic stress-induced pain remains limited. In this study, we investigated the function of lysine-specific histone demethylase 1A (KDM1A/LSD1) in the context of chronic overlapping pain comorbid with anxiety and depression in female mice. We employed a chronic variable stress model to induce pain hypersensitivity in the face and hindpaws, as well as anxiety-like and depression-like behaviors, in female mice. Our findings revealed that chronic stress led to a downregulation of KDM1A mRNA and protein expression in the mPFC. Notably, overexpressing KDM1A in the mPFC alleviated the pain hypersensitivity, anxiety-like behaviors, and depression-like behaviors in female mice, without affecting basal pain responses or inducing emotional distress. Conversely, conditional knockout of KDM1A in the mPFC exacerbated pain sensitivity and emotional distress specifically in females. In summary, this study highlights the crucial role of KDM1A in the mPFC in modulating chronic stress-induced overlapping pain, anxiety, and depression in females. Our findings suggest that KDM1A may serve as a potential therapeutic target for treating chronic stress-related overlap pain and associated negative emotional disorders.


Asunto(s)
Dolor Crónico , Regulación hacia Abajo , Histona Demetilasas , Ratones Endogámicos C57BL , Corteza Prefrontal , Estrés Psicológico , Animales , Corteza Prefrontal/metabolismo , Femenino , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Ratones , Dolor Crónico/metabolismo , Dolor Crónico/psicología , Depresión/metabolismo , Depresión/etiología , Ansiedad/metabolismo , Ratones Noqueados
9.
Cell Rep ; 43(5): 114230, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38743566

RESUMEN

Satellite glial cells (SGCs) of dorsal root ganglia (DRGs) are activated in a variety of chronic pain conditions; however, their mediation roles in pain remain elusive. Here, we take advantage of proteolipid protein (PLP)/creERT-driven recombination in the periphery mainly occurring in SGCs of DRGs to assess the role of SGCs in the regulation of chronic mechanical hypersensitivity and pain-like responses in two organs, the distal colon and hindpaw, to test generality. We show that PLP/creERT-driven hM3Dq activation increases, and PLP/creERT-driven TrkB.T1 deletion attenuates, colon and hindpaw chronic mechanical hypersensitivity, positively associating with calcitonin gene-related peptide (CGRP) expression in DRGs and phospho-cAMP response element-binding protein (CREB) expression in the dorsal horn of the spinal cord. Activation of Plp1+ DRG cells also increases the number of small DRG neurons expressing Piezo2 and acquiring mechanosensitivity and leads to peripheral organ neurogenic inflammation. These findings unravel a role and mechanism of Plp1+ cells, mainly SGCs, in the facilitation of chronic mechanical pain and suggest therapeutic targets for pain mitigation.


Asunto(s)
Dolor Crónico , Ganglios Espinales , Canales Iónicos , Neuronas , Regulación hacia Arriba , Animales , Ganglios Espinales/metabolismo , Dolor Crónico/metabolismo , Dolor Crónico/patología , Dolor Crónico/genética , Neuronas/metabolismo , Ratones , Canales Iónicos/metabolismo , Canales Iónicos/genética , Colon/metabolismo , Colon/patología , Masculino , Hiperalgesia/metabolismo , Hiperalgesia/patología , Proteína Proteolipídica de la Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Neuroglía/metabolismo
10.
Brain Behav Immun ; 119: 750-766, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38710336

RESUMEN

Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.


Asunto(s)
Dolor Crónico , Disbiosis , Ganglios Espinales , Microbioma Gastrointestinal , Ratones Noqueados , Miosina Tipo I , Animales , Femenino , Masculino , Ratones , Dolor Crónico/metabolismo , Dolor Crónico/microbiología , Disbiosis/metabolismo , Ganglios Espinales/metabolismo , Microbioma Gastrointestinal/fisiología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Miosina Tipo I/metabolismo
11.
Mol Brain ; 17(1): 22, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702738

RESUMEN

We previously reported that enhanced corticotropin-releasing factor (CRF) signaling in the bed nucleus of the stria terminalis (BNST) caused the aversive responses during acute pain and suppressed the brain reward system during chronic pain. However, it remains to be examined whether chronic pain alters the excitability of CRF neurons in the BNST. In this study we investigated the chronic pain-induced changes in excitability of CRF-expressing neurons in the oval part of the BNST (ovBNSTCRF neurons) by whole-cell patch-clamp electrophysiology. CRF-Cre; Ai14 mice were used to visualize CRF neurons by tdTomato. Electrophysiological recordings from brain slices prepared from a mouse model of neuropathic pain revealed that rheobase and firing threshold were significantly decreased in the chronic pain group compared with the sham-operated control group. Firing rate of the chronic pain group was higher than that of the control group. These data indicate that chronic pain elevated neuronal excitability of ovBNSTCRF neurons.


Asunto(s)
Dolor Crónico , Hormona Liberadora de Corticotropina , Neuronas , Núcleos Septales , Animales , Núcleos Septales/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Dolor Crónico/fisiopatología , Dolor Crónico/metabolismo , Masculino , Potenciales de Acción/fisiología , Ratones Endogámicos C57BL , Ratones
12.
Brain Behav Immun ; 120: 1-9, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38772429

RESUMEN

Aberrant neuronal excitability in the anterior cingulate cortex (ACC) is implicated in cognitive and affective pain processing. Such excitability may be amplified by activated circulating immune cells, including T lymphocytes, that interact with the central nervous system. Here, we conducted a study of individuals with chronic pain using magnetic resonance spectroscopy (MRS) to investigate the clinical evidence for the interaction between peripheral immune activation and prefrontal excitatory-inhibitory imbalance. In thirty individuals with chronic musculoskeletal pain, we assessed markers of peripheral immune activation, including soluble interleukin-2 receptor alpha chain (sCD25) levels, as well as brain metabolites, including Glx (glutamate + glutamine) to GABA+ (γ-aminobutyric acid + macromolecules/homocarnosine) ratio in the ACC. We found that the circulating level of sCD25 was associated with prefrontal Glx/GABA+. Greater prefrontal Glx/GABA+ was associated with higher pain catastrophizing, evaluative pain ratings, and anxiodepressive symptoms. Further, the interaction effect of sCD25 and prefrontal Glx/GABA+ on pain catastrophizing was significant, indicating the joint association of these two markers with pain catastrophizing. Our results provide the first evidence suggesting that peripheral T cellular activation, as reflected by elevated circulating sCD25 levels, may be linked to prefrontal excitatory-inhibitory imbalance in individuals with chronic pain. The interaction between these two systems may play a role as a potential mechanism underlying pain catastrophizing. Further prospective and treatment studies are needed to elucidate the specific role of the immune and brain interaction in pain catastrophizing.


Asunto(s)
Dolor Crónico , Subunidad alfa del Receptor de Interleucina-2 , Corteza Prefrontal , Humanos , Masculino , Femenino , Dolor Crónico/metabolismo , Corteza Prefrontal/metabolismo , Adulto , Persona de Mediana Edad , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/sangre , Giro del Cíngulo/metabolismo , Ácido Glutámico/metabolismo , Ácido Glutámico/sangre , Espectroscopía de Protones por Resonancia Magnética , Glutamina/metabolismo , Glutamina/sangre , Ácido gamma-Aminobutírico/metabolismo , Catastrofización/metabolismo
13.
Brain Behav Immun ; 119: 261-271, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38570102

RESUMEN

Upregulation of soluble tumor necrosis factor (sTNF) cytokine signaling through TNF receptor 1 (TNFR1) and subsequent neuronal hyperexcitability are observed in both animal models and human chronic neuropathic pain (CNP). Previously, we have shown that estrogen modulates sTNF/TNFR1 signaling in CNP, which may contribute to female prevalence of CNP. The estrogen-dependent role of TNFR1-mediated supraspinal neuronal circuitry in CNP remains unknown. In this study, we interrogated the intersect between supraspinal TNFR1 mediated neuronal signaling and sex specificity by selectively removing TNFR1 in Nex + neurons in adult mice (NexCreERT2::TNFR1f/f). We determined that mechanical hypersensitivity induced by chronic constriction injury (CCI) decreases over time in males, but not in females. Subsequently, we investigated two downstream pathways, p38MAPK and NF-κB, important in TNFR1 signaling and injury response. We detected p38MAPK and NF-κB activation in male cortical tissue; however, p38MAPK phosphorylation was reduced in NexCreERT2::TNFR1f/f males. We observed a similar recovery from acute pain in male mice following CCI when p38αMAPK was knocked out of supraspinal Nex + neurons (NexCreERT2::p38αMAPKf/f), while chronic pain developed in female mice. To explore the intersection between estrogen and inflammation in CNP we used a combination therapy of an estrogen receptor ß (ER ß) inhibitor with a sTNF/TNFR1 or general p38MAPK inhibitor. We determined both combination therapies lends therapeutic relief to females following CCI comparable to the response evaluated in male mice. These data suggest that TNFR1/p38αMAPK signaling in Nex + neurons in CNP is male-specific and lack of therapeutic efficacy following sTNF inhibition in females is due to ER ß interference. These studies highlight sex-specific differences in pathways important to pain chronification and elucidate potential therapeutic strategies that would be effective in both sexes.


Asunto(s)
Dolor Crónico , Estrógenos , Neuralgia , Neuronas , Receptores Tipo I de Factores de Necrosis Tumoral , Transducción de Señal , Animales , Neuralgia/metabolismo , Masculino , Femenino , Ratones , Estrógenos/metabolismo , Estrógenos/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Neuronas/metabolismo , Dolor Crónico/metabolismo , Transducción de Señal/fisiología , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Hiperalgesia/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Neural Transm (Vienna) ; 131(7): 739-753, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38630191

RESUMEN

Diagnosing and treating chronic orofacial pain is challenging due to its complex structure and limited understanding of its causes and mechanisms. In this study, we used RNA sequencing to identify differentially expressed genes (DEGs) in the rostral ventral medulla (RVM) and thalamus of rats with persistent orofacial pain, aiming to explore its development. DEGs were functionally analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Results showed a significant association between immune response and pain in this model. Key DEG mRNA expression trends were further validated using real-time quantitative polymerase chain reaction (RT-PCR), confirming their crucial roles in chronic orofacial pain. After injecting complete Freund's adjuvant (CFA) into the bilateral temporomandibular joint cavity for 14 days, we observed 293 upregulated genes and 14 downregulated genes in the RVM, and 1086 upregulated genes and 37 downregulated genes in the thalamus. Furthermore, we identified 27 common DEGs with altered expression (upregulation) in both the thalamus and RVM, including Cd74, C3, Cxcl13, C1qb, Itgal, Fcgr2b, C5ar1, and Tlr2, which are pain-associated genes. Protein-protein interaction (PPI) analysis using Cytoscape revealed the involvement of Toll-like receptors, complement system, differentiation clusters, and antigen presentation-related proteins in the interaction between the thalamus and RVM. The results of this study show that the immune system seems to have a more significant influence on chronic orofacial pain. There may be direct or indirect influence between the thalamus and RVM, which may participate in the regulation of chronic orofacial pain.


Asunto(s)
Dolor Crónico , Dolor Facial , Bulbo Raquídeo , Ratas Sprague-Dawley , Tálamo , Animales , Dolor Facial/genética , Dolor Facial/metabolismo , Dolor Facial/fisiopatología , Bulbo Raquídeo/metabolismo , Masculino , Ratas , Dolor Crónico/genética , Dolor Crónico/metabolismo , Tálamo/metabolismo , Análisis de Secuencia de ARN , Modelos Animales de Enfermedad , Transcriptoma
15.
J Integr Neurosci ; 23(4): 82, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38682225

RESUMEN

BACKGROUND: Comorbid chronic neuropathic pain (NPP) and anxio-depressive disorders (ADD) have become a serious global public-health problem. The SLIT and NTRK-like 1 (SLITRK1) protein is important for synaptic remodeling and is highly expressed in the amygdala, an important brain region involved in various emotional behaviors. We examined whether SLITRK1 protein in the amygdala participates in NPP and comorbid ADD. METHODS: A chronic NPP mouse model was constructed by L5 spinal nerve ligation; changes in chronic pain and ADD-like behaviors were measured in behavioral tests. Changes in SLITRK1 protein and excitatory synaptic functional proteins in the amygdala were measured by immunofluorescence and Western blot. Adeno-associated virus was transfected into excitatory synaptic neurons in the amygdala to up-regulate the expression of SLITRK1. RESULTS: Chronic NPP-related ADD-like behavior was successfully produced in mice by L5 ligation. We found that chronic NPP and related ADD decreased amygdalar expression of SLITRK1 and proteins important for excitatory synaptic function, including Homer1, postsynaptic density protein 95 (PSD95), and synaptophysin. Virally-mediated SLITRK1 overexpression in the amygdala produced a significant easing of chronic NPP and ADD, and restored the expression levels of Homer1, PSD95, and synaptophysin. CONCLUSION: Our findings indicated that SLITRK1 in the amygdala plays an important role in chronic pain and related ADD, and may prove to be a potential therapeutic target for chronic NPP-ADD comorbidity.


Asunto(s)
Amígdala del Cerebelo , Conducta Animal , Dolor Crónico , Homólogo 4 de la Proteína Discs Large , Proteínas del Tejido Nervioso , Neuralgia , Animales , Masculino , Ratones , Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Ansiedad/fisiopatología , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/fisiopatología , Conducta Animal/fisiología , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Depresión/metabolismo , Depresión/etiología , Depresión/fisiopatología , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Proteínas de Andamiaje Homer/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/metabolismo , Sinaptofisina/metabolismo
16.
Mol Immunol ; 170: 88-98, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38643689

RESUMEN

The pathogenesis of neuropathic pain (NP) is complex, and there are various pathological processes. Previous studies have suggested that lncRNA PCAT19 is abnormally expressed in NP conduction and affects the occurrence and development of pain. The aim of this study is to analyze the role and mechanism of PCAT19 in NP induced by chronic compressive nerve injury (CCI) in mice. In this study, C57BL/6 mice were applied to establish the CCI model. sh-PCAT19 was intrathecally injected once a day for 5 consecutive days from the second day after surgery. We discovered that PCat19 level was gradually up-regulated with the passage of modeling time. Downregulation of Iba-1-positive expression, M1/M2 ratio of microglia, and pro-inflammatory factors in the spinal cords of CCI-mice after PCat19 knock-downed was observed. Mechanically, the expression of miR-378a-3p was negatively correlated with KDM3A and PCat19. Deletion of KDM3A prevented H3K9me2 demethylation of BDNF promoter and suppressed BDNF expression. Further, KDM3A promotes CCI-induced neuroinflammation and microglia activation by mediating Brain-derived neurotrophic factor (BDNF) demethylation. Together, the results suggest that PCat19 may be involved in the development of NP and that PCat19 shRNA injection can attenuate microglia-induced neuroinflammation by blocking KDM3A-mediated demethylation of BDNF and BDNF release.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , MicroARNs , Microglía , Neuralgia , ARN Largo no Codificante , Animales , Masculino , Ratones , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Dolor Crónico/genética , Dolor Crónico/metabolismo , Desmetilación , Modelos Animales de Enfermedad , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , ARN Endógeno Competitivo , ARN Largo no Codificante/genética
17.
Autophagy ; 20(7): 1559-1576, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38522078

RESUMEN

A large proportion of patients with chronic pain experience co-morbid anxiety. The medial prefrontal cortex (mPFC) is proposed to underlie this comorbidity, but the molecular and neuronal mechanisms are not fully understood. Here, we reported that impaired neuronal macroautophagy in the prelimbic cortical (PrL) subregion of the mPFC paralleled the occurrence of anxiety-like behaviors in rats with chronic spared nerve injury (SNI). Intriguingly, such macroautophagy impairment was mainly observed in a FOS/c-Fos+ neuronal subpopulation in the PrL. Chemogenetic inactivation of this comorbid anxiety-related neuronal ensemble relieved pain-induced anxiety-like behaviors. Rescuing macroautophagy impairment in this neuronal ensemble relieved chronic pain-associated anxiety and mechanical allodynia and restored synaptic homeostasis at the molecular level. By contrast, artificial disruption of macroautophagy induced early-onset co-morbid anxiety in neuropathic rats, but not general anxiety in normal rats. Taken together, our work identifies causal linkage between PrL neuronal macroautophagy dysfunction and comorbid anxiety in neuropathic pain and provides novel insights into the role of PrL by differentiating its contribution in pain-induced comorbid anxiety from its modulation over general anxiety-like behaviors.Abbreviation: AAV: adeno-associated viruses; ACC: anterior cingulate cortex; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG12: autophagy related 12; CAMK2/CaMKII: calcium/calmodulin-dependent protein kinase II; CNO: clozapine-N-oxide; CQ: chloroquine; DIA: data independent acquisition; DIO: double floxed inverse orf; DLG4/PSD-95: discs large MAGUK scaffold protein 4; Dox: doxycycline; GABA: γ-aminobutyric acid; GFP: green fluorescent protein; GO: gene ontology; Gi: inhibitory guanine nucleotide-binding proteins; HsCHRM4/M4D: human cholinergic receptor muscarinic 4; HsSYN: human synapsin; KEGG: Kyoto encyclopedia of genes and genomes; LAMP1: lysosomal-associated membrane protein 1; LC3-II: PE conjugated microtubule-associated protein 1 light chain3; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mPFC: medial prefrontal cortex; P2A: 2A self-cleaving peptide; PPI: protein-protein interaction networks; PrL: prelimbic cortex; RBFOX3/NeuN: RNA binding protein, fox-1 homolog (C. elegans) 3; rtTA: reverse tetracycline-transactivator; SDS-PAGE: sodium dodecylsulfate-polyacrylamide gel electrophoresis; SHANK3: SH3 and multiple ankyrin repeat domains 3; SLC1A1/EAAC1: solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, systemXag), member 1; SNAP23: synaptosomal-associated protein 23; SNI:spared nerve injury; SQSTM1/p62: sequestosome 1; SYT3: synaptotagmin 3; TRE: tetracycline-responsive element; TRE3G: third-generation tetracycline-responsive element.


Asunto(s)
Ansiedad , Macroautofagia , Neuralgia , Neuronas , Corteza Prefrontal , Animales , Neuralgia/metabolismo , Corteza Prefrontal/metabolismo , Ratas , Neuronas/metabolismo , Masculino , Macroautofagia/fisiología , Ratas Sprague-Dawley , Conducta Animal , Dolor Crónico/metabolismo , Autofagia/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hiperalgesia
18.
J Neurosci ; 44(16)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38453466

RESUMEN

Chronic pain and alcohol use disorder (AUD) are highly comorbid, and patients with chronic pain are more likely to meet the criteria for AUD. Evidence suggests that both conditions alter similar brain pathways, yet this relationship remains poorly understood. Prior work shows that the anterior insular cortex (AIC) is involved in both chronic pain and AUD. However, circuit-specific changes elicited by the combination of pain and alcohol use remain understudied. The goal of this work was to elucidate the converging effects of binge alcohol consumption and chronic pain on AIC neurons that send projections to the dorsolateral striatum (DLS). Here, we used the Drinking-in-the-Dark (DID) paradigm to model binge-like alcohol drinking in mice that underwent spared nerve injury (SNI), after which whole-cell patch-clamp electrophysiological recordings were performed in acute brain slices to measure intrinsic and synaptic properties of AIC→DLS neurons. In male, but not female, mice, we found that SNI mice with no prior alcohol exposure consumed less alcohol compared with sham mice. Electrophysiological analyses showed that AIC→DLS neurons from SNI-alcohol male mice displayed increased neuronal excitability and increased frequency of miniature excitatory postsynaptic currents. However, mice exposed to alcohol prior to SNI consumed similar amounts of alcohol compared with sham mice following SNI. Together, our data suggest that the interaction of chronic pain and alcohol drinking have a direct effect on both intrinsic excitability and synaptic transmission onto AIC→DLS neurons in mice, which may be critical in understanding how chronic pain alters motivated behaviors associated with alcohol.


Asunto(s)
Alcoholismo , Consumo Excesivo de Bebidas Alcohólicas , Dolor Crónico , Enfermedades del Sistema Nervioso Periférico , Humanos , Ratones , Animales , Masculino , Dolor Crónico/metabolismo , Corteza Insular , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Etanol/farmacología , Neuronas/metabolismo , Alcoholismo/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo
19.
J Nanobiotechnology ; 22(1): 128, 2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38519978

RESUMEN

Accumulating evidence supports the notion that microglia play versatile roles in different chronic pain conditions. However, therapeutic strategies of chronic pain by targeting microglia remain largely overlooked. This study seeks to develop a miRNA-loaded nano-delivery system by targeting microglia, which could provide a decent and long-lasting analgesia for chronic pain. Surface aminated mesoporous silica nanoparticles were adopted to load miR-26a-5p, a potent analgesic miRNA, by electrostatic adsorption, which can avoid miR-26a-5p is rapidly released and degraded. Then, targeting peptide MG1 was modified on the surface of aminated mesoporous silica particles for microglia targeting. In peripheral nerve injury induced neuropathic pain model, a satisfactory anti-allodynia effect with about 6 weeks pain-relief duration were achieved through targeting microglia strategy, which decreased microglia activation and inflammation by Wnt5a, a non-canonical Wnt pathway. In inflammatory pain and chemotherapy induced peripheral neuropathic pain, microglia targeting strategy also exhibited more efficient analgesia and longer pain-relief duration than others. Overall, we developed a microglia-targeting nano-delivery system, which facilitates precisely miR-26a-5p delivery to enhance analgesic effect and duration for several chronic pain conditions.


Asunto(s)
Analgesia , Dolor Crónico , MicroARNs , Nanopartículas , Neuralgia , Humanos , Microglía/metabolismo , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , MicroARNs/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/genética , Neuralgia/metabolismo , Analgésicos/metabolismo , Analgésicos/farmacología , Analgésicos/uso terapéutico , Dióxido de Silicio/farmacología
20.
J Cell Mol Med ; 28(4): e18136, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38334255

RESUMEN

Chronic pain is the key manifestations of rheumatoid arthritis. Neuroinflammation in the spinal cord drives central sensitization and chronic pain. Ferroptosis has potentially important roles in the occurrence of neuroinflammation and chronic pain. In the current study, mouse model of collagen-induced arthritis was established by intradermal injection of type II collagen in complete Freund's adjuvant (CFA) solution. CFA inducement resulted in swollen paw and ankle, mechanical and spontaneous pain, and impaired motor coordination. The spinal inflammation was triggered, astrocytes were activated, and increased NLRP3-mediated inflammatory signal was found in CFA spinal cord. Oxidative stress and ferroptosis in the spinal cord were manifested. Meanwhile, enhancive spinal GSK-3ß activity and abnormal phosphorylated Drp1 were observed. To investigate the potential therapeutic options for arthritic pain, mice were intraperitoneally injected with AB4 for three consecutive days. AB4 treatment reduced pain sensitivity and increased the motor coordination. In the spinal cord, AB4 treatment inhibited NLRP3 inflammasome-mediated inflammatory response, increased antioxidation, decreased mitochondrial reactive oxygen species and ferroptosis. Furthermore, AB4 decreased GSK-3ß activity by binding with GSK-3ß through five electrovalent bonds. Our findings indicated that AB treatment relieves arthritis pain by inhibiting GSK-3ß activation, increasing antioxidant capability, reducing Drp1-mediated mitochondrial dysfunction and suppressing neuroinflammation.


Asunto(s)
Artritis Reumatoide , Dolor Crónico , Ferroptosis , Saponinas , Ratones , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Dolor Crónico/metabolismo , Enfermedades Neuroinflamatorias , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA