Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pain ; 20: 17448069241242982, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38485252

RESUMEN

Itch is a somatosensory sensation to remove potential harmful stimulation with a scratching desire, which could be divided into mechanical and chemical itch according to diverse stimuli, such as wool fiber and insect biting. It has been reported that neuropeptide Y (NPY) neurons, a population of spinal inhibitory interneurons, could gate the transmission of mechanical itch, with no effect on chemical itch. In our study, we verified that chemogenetic activation of NPY neurons could inhibit the mechanical itch as well as the chemical itch, which also attenuated the alloknesis phenomenon in the chronic dry skin model. Afterwards, intrathecal administration of NPY1R agonist, [Leu31, Pro34]-NPY (LP-NPY), showed the similar inhibition effect on mechanical itch, chemical itch and alloknesis as chemo-activation of NPY neurons. Whereas, intrathecal administration of NPY1R antagonist BIBO 3304 enhanced mechanical itch and reversed the alloknesis phenomenon inhibited by LP-NPY treatment. Moreover, selectively knocking down NPY1R by intrathecal injection of Npy1r siRNA enhanced mechanical and chemical itch behavior as well. These results indicate that NPY neurons in spinal cord regulate mechanical and chemical itch, and alloknesis in dry skin model through NPY1 receptors.


Asunto(s)
Neuropéptido Y , Receptores de Neuropéptido Y , Animales , Prurito/inducido químicamente , Transducción de Señal , Médula Espinal
2.
Exp Cell Res ; 418(1): 113224, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35643178

RESUMEN

PURPOSE: Acute respiratory distress syndrome (ARDS), a severe medical condition, is among the major causes of death in critically ill patients. Morphine is used as a therapeutic agent against severe pain. The mechanisms of its reactions over ARDS are not fully understood. The aim of this study was to assess the mechanism of morphine in rats with ARDS. METHODS: Rats were injected with lipopolysaccharide to induce ARDS, and some rats were pre-treated with graded doses of morphine in the lateral ventricles to assess survival and non-infected mortality. Immunohistochemical and HE staining were performed to measure MPO and CD68 activity in the lungs and lung injury. ELISA was conducted to detect the inflammatory factor levels in the plasma and BALF. Co-labeling of µ-opioid receptor (MOR) and c-Fos was observed in the brain tissues. MOR-positive cells in brain tissues were evaluated using immunohistochemistry. The effect of MOR antagonists on ARDS was examined in rats by pre-injection of naloxone or methylnaltrexone. The expression of MyD88, TLR4, and NF-κB was lastly assessed. RESULTS: Dose-independent improvement was observed in respiratory capacity and lung injury in ARDS rats after morphine pre-injection, along with reduced inflammatory factors in the plasma and BALF. MOR-positive cells were elevated after morphine, which occurred within the ventral part of the gigantocellular reticular nucleus (GiV). Naloxone and methylnaltrexone blocked the effects of morphine via central and peripheral MOR. Morphine activated TLR pathway in a MyD88-dependent manner. CONCLUSION: Morphine activates MOR within the GiV and the TLR pathway to attenuate ARDS in rats.


Asunto(s)
Lesión Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Lipopolisacáridos , Morfina/farmacología , Factor 88 de Diferenciación Mieloide , Naloxona/farmacología , Ratas , Receptores Opioides , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
3.
J Neurosci ; 41(37): 7727-7741, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34349001

RESUMEN

Chronic itch is a troublesome condition and often difficult to cure. Emerging evidence suggests that the periaqueductal gray (PAG)-rostral ventromedial medulla (RVM) pathway may play an important role in the regulation of itch, but the cellular organization and molecular mechanisms remain incompletely understood. Here, we report that a group of RVM neurons distinctively express the G-protein-coupled estrogen receptor (GPER), which mediates descending inhibition of itch. We found that GPER+ neurons in the RVM were activated in chronic itch conditions in rats and mice. Selective ablation or chemogenetic suppression of RVM GPER+ neurons resulted in mechanical alloknesis and increased scratching in response to pruritogens, whereas chemogenetic activation of GPER+ neurons abrogated itch responses, indicating that GPER+ neurons are antipruritic. Moreover, GPER-deficient mice and rats of either sex exhibited hypersensitivity to mechanical and chemical itch, a phenotype reversible by the µ type opioid receptor (MOR) antagonism. Additionally, significant MOR phosphorylation in the RVM was detected in chronic itch models in wild-type but not in GPER-/- rats. Therefore, GPER not only identifies a population of medullary antipruritic neurons but may also determine the descending antipruritic tone through regulating µ opioid signaling.SIGNIFICANCE STATEMENT Therapeutic options for itch are limited because of an as yet incomplete understanding of the mechanisms of itch processing. Our data have provided novel insights into the cellular organization and molecular mechanisms of descending regulation of itch in normal and pathologic conditions. GPER+ neurons (largely GABAergic) in the RVM are antipruritic neurons under tonic opioidergic inhibition, activation of GPER promotes phosphorylation of MOR and disinhibition of the antipruritic GPER+ neurons from inhibitory opioidergic inputs, and failure to mobilize GPER+ neurons may result in the exacerbation of itch. Our data also illuminate on some of the outstanding questions in the field, such as the mechanisms underlying sex bias in itch, pain, and opioid analgesia and the paradoxical effects of morphine on pain and itch.


Asunto(s)
Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Femenino , Masculino , Ratones , Fosforilación , Prurito/genética , Prurito/metabolismo , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Receptores Opioides mu/metabolismo , Transducción de Señal/fisiología
4.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 404-416, 2022 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-36514220

RESUMEN

The dry skin tortures numerous patients with severe itch. The transient receptor potential cation channel V member 1 (TRPV1) and A member 1 (TRPA1) are two essential receptors for peripheral neural coding of itch sensory, mediating histaminergic and nonhistaminergic itch separately. In the dorsal root ganglion, transmembrane protein 100 (TMEM100) is structurally related to both TRPV1 and TRPA1 receptors, but the exact role of TMEM100 in itch sensory coding is still unknown. Here, in this study, we find that TMEM100 + DRG neurons account for the majority of activated neurons in an acetone-ether-water (AEW)-induced dry skin itch model, and some TMEM100 + DRG neurons are colocalized with both TRPA1 and the chloroquine-related Mrgpr itch receptor family. Both the expression and function of TRPA1 channels, but not TRPV1 channels, are upregulated in the AEW model, and specific DRG Tmem100 gene knockdown alleviates AEW-induced itch and rescues the expression and functional changes of TRPA1. Our results strongly suggest that TMEM100 protein in DRG is the main facilitating factor for dry-skin-related chronic itch, and specific suppression of TMEM100 in DRG could be a novel effective treatment strategy for patients who suffer from dry skin-induced itch.


Asunto(s)
Prurito , Canales de Potencial de Receptor Transitorio , Humanos , Ganglios Espinales/metabolismo , Proteínas de la Membrana/metabolismo , Prurito/inducido químicamente , Prurito/genética , Prurito/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo , Canal Catiónico TRPA1/genética , Canal Catiónico TRPA1/metabolismo , Regulación hacia Arriba
5.
Biochem Biophys Res Commun ; 557: 69-76, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33862462

RESUMEN

Remifentanil is a potent, short-acting opioid analgesic drug that can protect tissues from ischemia and reperfusion injury though anti-inflammatory effects. However, the utility of remifentanil in liver regeneration after hepatectomy is not known. Using a 70% hepatectomy mouse model (PHx), we found that preconditioning animals with 4 µg/kg remifentanil enhanced liver regeneration through supporting hepatocyte proliferation but not through anti-inflammatory effects. These effects were also phenocopied in vitro where 40 mM remifentanil promoted the proliferation of primary mouse hepatocyte cultures. We further identified that remifentanil treatment increased the expression of ß-arrestin 2 in vivo and in vitro. Demonstrating specificity, remifentanil preconditioning failed to promote liver regeneration in liver-specific ß-arrestin 2 knockout (CKO) mice subjected to PHx. While remifentanil increased the expression of activated (phosphorylated)-ERK and cyclin D1 in PHx livers, their levels were not significantly changed in remifentanil-treated CKO mice nor in WT mice pretreated with the ERK inhibitor U0126. Our findings suggest that remifentanil promotes liver regeneration via upregulation of a ß-arrestin 2/ERK/cyclin D1 axis, with implications for improving regeneration process after hepatectomy.


Asunto(s)
Ciclina D1/metabolismo , Regeneración Hepática , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Remifentanilo/farmacología , Daño por Reperfusión/terapia , Arrestina beta 2/metabolismo , Analgésicos Opioides/farmacología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Hepatectomía , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Regulación hacia Arriba
6.
Arch Toxicol ; 95(7): 2431-2442, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33852043

RESUMEN

Hepatocytes are the main cell components of the liver and perform metabolic, detoxification, and endocrine functions. Functional hepatocytes are of great value in drug development, toxicity evaluation, and cell therapy for liver diseases. In recent years, an increasing number of in vitro models have been developed to screen drugs and test their toxicity. However, maintaining hepatocyte function in vitro for a long time is a serious challenge. Even freshly isolated liver cells cultured for a short time may lose function via spontaneous dedifferentiation. Thus, novel cell culture systems allowing extended hepatocyte maintenance and more predictive long-term in vitro studies are required. In this study, we developed a conditioned culture system composed of a small-molecule combination that can maintain hepatocyte morphology and functions over the long term. Two-month culture of primary human hepatocytes showed that the conditioned medium was able to stably preserve hepatic functions such as albumin and α-antitrypsin secretion, hepatic transport activity, urea synthesis, and ammonia elimination. Furthermore, this culture model can be used to assess drug-induced hepatotoxicity in vitro. In summary, our work suggests a feasible approach to maintain hepatocyte function in vitro and proposes a promising model for long-term toxicological studies and drug development.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Hepatopatías , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Hepatopatías/metabolismo
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1177-1188, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34244711

RESUMEN

Stroke is the second leading cause of death and long-term disability worldwide, which lacks effective treatment. Perioperative stroke is associated with much higher rates of mortality and disability. The neuroprotective role of dexmedetomidine (Dex), a highly selective agonist of alpha2-adrenergic receptor, has been reported in a stroke rat model, and it was found that pretreatment of Dex before stroke could alleviate blood-brain barrier (BBB) breakdown. However, the underlying mechanisms are still unknown. As the brain endothelial cells are the main constituents of BBB and in high demand of energy, mitochondrial function of endothelial cells plays an important role in the maintenance of BBB. Given that dynamin-related protein 1 (Drp1) is a protein mediating mitochondrial fission, with mitochondrial fusion that balances mitochondrial morphology and ensures mitochondria function, the present study was designed to investigate the possible role of Drp1 in endothelial cells involved in the neuroprotective effects of Dex in ischemic stroke. Our results showed that preconditioning with Dex reduced infarction volume, alleviated brain water content and BBB damage, and improved neurological scores in middle cerebral artery occlusion rats. Meanwhile, Dex enhanced cell activity and decreased cell apoptosis in oxygen-glucose deprivation human brain microvascular endothelial cells in vitro. These protective effects of Dex were correlated with the mitochondrial morphology integrality of endothelial cells, mediated by increased phosphorylation of serine 637 in Drp1, and could be reversed by α2-adrenergic receptor antagonist Yohimbine and AMP-activated protein kinase inhibitor Compound C. These findings suggest new molecular pathways involved in the neuroprotective effects of Dex in ischemic stroke. As Dex is routinely used as a sedative drug clinically, our findings provide molecular evidence that it has perioperative neuroprotection from ischemic stroke.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Dexmedetomidina/farmacología , Dinaminas/metabolismo , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Dexmedetomidina/uso terapéutico , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Alcaloides Indólicos/farmacología , Alcaloides Indólicos/uso terapéutico , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Accidente Cerebrovascular Isquémico/etiología , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Dinámicas Mitocondriales/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Proteína de la Zonula Occludens-1/metabolismo
8.
Acta Biochim Biophys Sin (Shanghai) ; 53(8): 1076-1087, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34137445

RESUMEN

Propofol is the most commonly used intravenous anesthetic worldwide. It can induce loss of consciousness prior to the occurrence of severe respiratory suppression, which is also a pharmacodynamic feature of all general anesthetics. However, the neural mechanisms underlying this natural phenomenon are controversial and highly related to patient safety. In the present study, we demonstrated that the pharmacodynamic effects of propofol (50 and 100 µM) on suppression of consciousness-related excitatory postsynaptic currents in the medial prefrontal cortex (mPFC) and centromedian nucleus of the thalamus (CMT) were lower than those in the kernel respiratory rhythmogenesis nucleus pre-Bötzinger complex (PrBo). Furthermore, we unexpectedly found that the GABAA receptor ß3 subunit is the key target for propofol's action and that it is mutually and exclusively expressed in GABAergic neurons. It is also more abundant in the mPFC and CMT, but mainly co-localized with GABAergic neurons in the PrBo. As a result, the differentiated expression pattern should mediate more neuron suppression through the activation of GABAergic neurons in the mPFC and CMT at low doses of propofol (50 µM). However, PrBo GABAergic neurons were only activated by propofol at a high dose (100 µM). These results highlight the detailed pharmacodynamic effects of propofol on consciousness-related and respiration-related nuclei and provide the distinct interaction mechanism between the ß3 subunit and GABAergic neurons in mediating the suppression of consciousness compared to the inhibition of respiration.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Núcleos Talámicos Intralaminares , Corteza Prefrontal , Propofol/farmacología , Receptores de GABA-A/metabolismo , Mecánica Respiratoria/efectos de los fármacos , Inconsciencia , Animales , Núcleos Talámicos Intralaminares/metabolismo , Núcleos Talámicos Intralaminares/fisiopatología , Masculino , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Ratas , Ratas Sprague-Dawley , Inconsciencia/inducido químicamente , Inconsciencia/metabolismo , Inconsciencia/fisiopatología
9.
Acta Biochim Biophys Sin (Shanghai) ; 53(7): 883-892, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-33929026

RESUMEN

Propofol is widely used for the induction and maintenance of anesthesia, which causes a rapid loss of consciousness. However, the mechanisms underlying the hypnosis effect of propofol are still not fully understood. The thalamic reticular nucleus (TRN) is crucial for regulating wakefulness, sleep rhythm generation, and sleep stability, while the role of TRN in the process of propofol-induced anesthesia is still unknown. Here, we investigated the function of the anterior TRN in propofol general anesthesia. Our results demonstrated that the neural activity of anterior TRN is suppressed during propofol anesthesia, whereas it is robustly activated from anesthesia by recording the calcium signals using fiber photometry technology. The results showed that the activation of anterior TRN neurons by chemogenetic and optogenetic methods shortens the emergency time without changing the induction time. Conversely, chemogenetic or optogenetic inhibition of the TRN neurons leads to a delay in the recovery time. Our study showed that anterior TRN is crucial for behavioral arousal without affecting the induction time of propofol anesthesia.


Asunto(s)
Núcleos Talámicos Anteriores/metabolismo , Nivel de Alerta/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Propofol/farmacología , Animales , Masculino , Ratones
10.
Acta Biochim Biophys Sin (Shanghai) ; 53(5): 538-546, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33693534

RESUMEN

Peripheral inflammation is always accompanied by a noxious sensation, either pain or itch, providing a protective warning for the occurrence of pathological changes; however, the mechanisms determining whether pain, itch, or both will be elicited under certain inflammatory statuses are still far from clear. Complete Freund's adjuvant (CFA) contains heat killed and dried Mycobacterium tuberculosis widely used to induce inflammatory pain models, but how CFA treatment affects itch sensation and the possible mechanisms are still unclear. In this study, using itch behavior testing and calcium imaging, we showed that both the behaviors and calcium responses associated with Transient Receptor Potential Vanilloid 1 (TRPV1)-mediated histamine-dependent itch and Transient Receptor Potential Ankyrin 1 (TRPA1)-mediated histamine-independent itch were significantly suppressed by CFA treatment. Furthermore, to explore the possible cellular mechanisms, high-throughput single-cell RNA sequencing and real-time PCR were used to detect CFA-induced changes of itch-related genes in dorsal root ganglion (DRG) neurons. Our results revealed that although both nociceptive Trpv1+ and Trpa1+ DRG neurons were increased after CFA treatment, most known pruriceptors, including Hrh1+, Mrgpra3+, Mrgprd+, Htr3a+, Htr1f+, IL31ra+, Osmr+, and Lpar3+ DRG neurons, were significantly decreased, which may explain that CFA treatment caused itch suppression. This study indicated that itch sensation was affected after CFA treatment, although negatively, and comprehensive but not specific suppression of different pruriceptors was observed after CFA treatment, suggesting that a unified adaptive change of increased pain and decreased itch will occur simultaneously under CFA-induced inflammatory conditions.


Asunto(s)
Adyuvante de Freund/farmacocinética , Prurito/tratamiento farmacológico , Canal Catiónico TRPA1/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Masculino , Ratones , Prurito/metabolismo , Prurito/patología
11.
Artículo en Inglés | MEDLINE | ID: mdl-32087972

RESUMEN

In both normal turnover of the hepatic tissue and acute hepatic injury, the liver predominantly activates terminally differentiated hepatocytes to proliferate and repair. However, in chronic and severe chronic injury, this capacity fails, and liver progenitor cells (LPCs) can give rise to hepatocytes to restore both hepatic architecture and liver metabolic function. Although the promotion of LPC-to-hepatocyte differentiation to acquire a considerable number of functional hepatocytes could serve as a potentially new therapeutic option for patients with end-stage liver disease, its development first requires the identification of the molecular mechanisms driving this process. Here, we found that the epithelial cell adhesion molecule (EpCAM), a progenitor cell marker, regulates the differentiation of LPCs into hepatocytes through Notch1 signaling pathway. Western blotting (WB) revealed a consistent expression pattern of EpCAM and Notch1 during LPC-to-hepatocyte differentiation in vitro. Additionally, overexpression of EpCAM blocked LPC-to-hepatocyte differentiation, which was in consistent with the repressive role of Notch signaling during hepatic differentiation. WB and immunofluorescence data also showed that the upregulation of EpCAM expression increased the generation of Notch intracellular domain (N1ICD), indicating the promotion of Notch1 activity. Our results established the EpCAM-Notch1 signaling axis as an inhibitory mechanism preventing LPC-to-hepatocyte differentiation in vitro.

12.
Mediators Inflamm ; 2020: 6458791, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32565727

RESUMEN

Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2/B1) is a protein involved in the regulation of RNA processing, cell metabolism, migration, proliferation, and apoptosis. However, the effect of hnRNPA2/B1 on injured endothelial cells (ECs) remains unclear. We investigated the effect of hnRNPA2/B1 on lipopolysaccharide- (LPS-) induced vascular endothelial injury in human umbilical vein endothelial cells (HUVECs) and the underlying mechanisms. LPS was used to induce EC injury, and the roles of hnRNPA2/B1 in EC barrier dysfunction and inflammatory responses were measured by testing endothelial permeability and the expression of inflammatory factors after the suppression and overexpression of hnRNPA2/B1. To explore the underlying mechanism by which hnRNPA2/B1 regulates endothelial injury, we studied the VE-cadherin/ß-catenin pathway and NF-κB activation in HUVECs. The results showed that hnRNPA2/B1 was elevated in LPS-stimulated HUVECs. Moreover, knockdown of hnRNPA2/B1 aggravated endothelial injury by increasing EC permeability and promoting the secretion of the inflammatory cytokines TNF-α, IL-1ß, and IL-6. Overexpression of hnRNPA2/B1 can reduce the permeability and inflammatory response of HUVEC stimulated by LPS in vitro, while increasing the expression of VE-Cadherin and ß-catenin. Furthermore, the suppression of hnRNPA2/B1 increased the LPS-induced NF-κB activation and reduced the VE-cadherin/ß-catenin pathway. Taken together, these results suggest that hnRNPA2/B1 can regulate LPS-induced EC damage through regulating the NF-κB and VE-cadherin/ß-catenin pathways.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Lipopolisacáridos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Apoptosis , Proliferación Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/metabolismo , Microscopía Fluorescente , Permeabilidad , ARN Interferente Pequeño/metabolismo
13.
Acta Biochim Biophys Sin (Shanghai) ; 52(8): 864-874, 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32515467

RESUMEN

During the rapidly developing and sensitive period of the central nervous system (CNS), a harmful stimulus may have serious consequences. The effect of anesthetic exposure on the development of the offspring's CNS during pregnancy is still unclear and has been widely concerned. In the present study, we compared the susceptibility of the hippocampus with those of other brain regions in offsprings when the mother mice were exposed to repeated sevoflurane. We found that other than affecting motor sensation, emotion, or social behavior of offspring mice, repeated sevoflurane exposure induced significant memory deficiency. Compared with other brain regions, the hippocampus, which is the key component of the brain serving for learning and memory, was more vulnerable to repeated sevoflurane exposure. We also found that repeated sevoflurane exposure to mother mice could inhibit the axon development of hippocampal neurons. We also predicted that N6-methyladenosine modification of mRNA might play an essential role in the vulnerability of the hippocampus to sevoflurane, while the underlying cellular mechanism needs to be explored in the future. Our study may provide a new perspective for studying the mechanism of hippocampus-specific injury induced by sevoflurane exposure.


Asunto(s)
Hipocampo , Exposición Materna/efectos adversos , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Sevoflurano/efectos adversos , Animales , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Sevoflurano/farmacología
14.
Neural Plast ; 2020: 8866187, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908490

RESUMEN

Ample evidence suggests that estrogens have strong influences on the occurrence of stress-related mood disorders, but the underlying mechanisms remain poorly understood. Through multiple approaches, we demonstrate that the G protein-coupled estrogen receptor (GPER) is widely distributed along the HPA axis and in brain structures critically involved in mood control. Genetic ablation of GPER in the rat resulted in significantly lower basal serum corticosterone level but enhanced ACTH release in response to acute restraint stress, especially in the female. GPER-/- rats of either sex displayed increased anxiety-like behaviors and deficits in learning and memory. Additionally, GPER deficiency led to aggravation of anxiety-like behaviors following single-prolonged stress (SPS). SPS caused significant decreases in serum corticosterone in WT but not in GPER-deficient rats. The results highlight an important role of GPER at multiple sites in regulation of the HPA axis and mood.


Asunto(s)
Ansiedad/sangre , Ansiedad/fisiopatología , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/fisiopatología , Sistema Hipófiso-Suprarrenal/fisiopatología , Receptores Acoplados a Proteínas G/fisiología , Hormona Adrenocorticotrópica/sangre , Animales , Conducta Animal , Femenino , Técnicas de Inactivación de Genes , Hipocampo/fisiología , Masculino , Ratas Transgénicas
15.
Acta Biochim Biophys Sin (Shanghai) ; 51(12): 1216-1222, 2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31735968

RESUMEN

The hypothalamus-pituitary-adrenal (HPA) axis is known to mediate gut-brain interaction, and the pathological inflammatory process in the intestine can induce HPA axis involved 'fight or flight' response to suppress or facilitate intestinal inflammation. Hypothalamic paraventricular nucleus (PVN) neurons are responsible for controlling the HPA axis activity, but their exact role in modulating intestinal inflammation remains unclear. In this study, we used the dextran sulfate sodium (DSS)-induced mice colitis model, gene editing, and RNA interference to determine the effects of PVN neurons on intestinal inflammation. We found that at the early stage (third day) after DSS treatment, there was a mild inflammation in the colorectal area and an increased neuron activation in the PVN but not in the adjacent area. At the same time, ~80% of activated PVN neurons also expressed novel estrogen GPER1 receptor. The colitis noticeably worsened in GPER1-knockout mice and local PVN GPER1-knockdown mice. These results indicated that PVN GPER1 positive neurons potentially have a protective function during the early stages of DSS-induced colitis, and this may be a mechanism by which the central nervous system attempts to suppress intestinal inflammation to achieve self-protection.


Asunto(s)
Colitis/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley
16.
Lab Invest ; 98(12): 1588-1599, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30089853

RESUMEN

Interleukin (IL)-18 plays an important role in liver ischemia/reperfusion (I/R) injury. We have previously demonstrated that remifentanil protects against liver I/R injury by upregulating the hepatic expression of IL-18-binding protein (IL-18BP), a natural IL-18 inhibitor. The current study was performed to further clarify the effects of remifentanil on IL-18BP expression in the liver as well as investigate the underlying mechanisms. In Sprague-Dawley (SD) rats, we demonstrated that remifentanil significantly increased the expression of IL-18BP in normal rat liver tissue over a 24-h time period with maximal expression at 24 h after treatment. The upregulation of remifentanil on IL-18BP expression displayed similar trends in in vitro cellular studies, including mouse primary hepatocytes, normal human hepatocyte LO2, and mouse hepatoma cells Hep1-6. In LO2 cells, preexposure of the cells to remifentanil significantly inhibited IL-18-activated p65 NF-κB phosphorylation, and the inhibition was absent when the cells were transfected with IL-18BP siRNA, indicating the functional effects of IL-18BP induced by remifentanil. Pretreatment with actinomycin D abolished remifentanil-induced upregulation of IL-18BP mRNA, suggesting that the induction occurred at the transcriptional level. This was further supported by the luciferase reporter assay, which demonstrated that remifentanil treatment significantly increased transcription of the IL-18BP promoter. Both western blot analysis and ChIP assays showed that STAT1 and C/EBP ß were activated by remifentanil. Furthermore, remifentanil failed to upregulate IL-18BP expression after silencing STAT1 or C/EBP ß gene expression. These findings demonstrate that remifentanil could upregulate hepatic IL-18BP expression through transcriptional activation of the IL-18BP promoter, and STAT1 and C/EBP ß are two key transcriptional factors involved in this process.


Asunto(s)
Analgésicos Opioides/farmacología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Hepatocitos/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Remifentanilo/farmacología , Factor de Transcripción STAT1/metabolismo , Animales , Línea Celular , Hepatocitos/metabolismo , Humanos , Hepatopatías/prevención & control , Masculino , Ratones , Ratas Sprague-Dawley , Daño por Reperfusión/prevención & control , Regulación hacia Arriba
17.
Cell Biol Int ; 42(10): 1311-1320, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29907991

RESUMEN

Aberrant aggregation and activation of lung fibroblasts is a key process in pulmonary fibrosis, but the underlying mechanism remains enigmatic. Forkhead Box O3a (FoxO3a) is considered to be an important transcription factor that could regulate both cell cycle and cell viability. To investigate the role of FoxO3a on LPS-induced lung fibroblast proliferation, we transfected FoxO3a-SiRNA or FoxO3a-OE lentivirus into cultured mouse lung fibroblasts to knockdown or overexpress FoxO3a and pretreated mouse lung fibroblasts with gefitinib to enhance FoxO3a activity. The proliferation of lung fibroblasts was evaluated by CCK8 assay, the expression of FoxO3a, phosphorylated FoxO3a (p-FoxO3a) and p27 were measured by Western blot. We found that the proliferation of mouse lung fibroblasts mediated by LPS is accompanied by the inactivation of FoxO3a. The knockdown of FoxO3a could further decreased the expression of p27 mediated by LPS, while the overexpression of FoxO3a significantly increased the expression of p27 and suppressed LPS-induced lung fibroblast proliferation. Upon treating fibroblasts with gefitinib, the phosphorylation of FoxO3a was reduced and FoxO3a translocated into the nucleus, the expression of p27 was significantly increased and the proliferation of lung fibroblasts mediated by LPS could also be inhibited effectively. The results indicate that overexpression and reduced phosphatase activity of FoxO3a inhibit LPS-induced lung fibroblast proliferation through the activation of FoxO3a/p27 signaling pathways. Thus, to enhance FoxO3a activity could be a potential therapeutic target for LPS-induced pulmonary fibrosis.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína Forkhead Box O3/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/citología , Proteína Forkhead Box O3/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Lipopolisacáridos/farmacología , Pulmón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Mediators Inflamm ; 2018: 3260256, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29861656

RESUMEN

Remifentanil, an ultra-short acting opiate, has been reported to protect against hepatic ischemia-reperfusion injury, which is a major cause of postoperative liver dysfunction. The objective of this study was to determine whether a central vagal pathway is involved in this protective procedure. Rat models of hepatic ischemia-reperfusion were used in the experimental procedures. The results revealed that intravenous pretreatment with remifentanil decreased serum aminotransferases and hepatic histologic damage; however, an intraperitoneal injection of µ-opioid receptor antagonist did not abolish the protection of remifentanil preconditioning. c-Fos immunofluorescence of the brain stem showed that dorsal motor nucleus of the vagus was activated after remifentanil preconditioning. Moreover, serum alanine aminotransferase, histopathologic damage, and apoptosis decreased in remifentanil preconditioning group compared to vagotomized animals with remifentanil preconditioning, and there was no statistical difference of TNF-α and IL-6 between NS/Va and RPC/Va groups. In addition, remifentanil microinjection into dorsal vagal complex decreased serum aminotransferases, inflammatory cytokines, and hepatic histologic injury and apoptosis, and these effects were also abolished by a peripheral hepatic vagotomy. In conclusion, remifentanil preconditioning conferred liver protection against ischemia-reperfusion injury, which was mediated by the central vagal pathway.


Asunto(s)
Piperidinas/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Alanina Transaminasa/sangre , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasas/sangre , Interleucina-6/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Remifentanilo , Daño por Reperfusión/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Nervio Vago
19.
Acta Biochim Biophys Sin (Shanghai) ; 50(12): 1219-1226, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30339176

RESUMEN

CC chemokine ligand 2 (CCL2) has been implicated in pathological pain, but the mechanism underlying the pronociceptive effect of CCL2 is not fully understood. Voltage-gated sodium (Nav) channels are important determinants of the excitability of sensory neurons. Hence we tested the hypothesis that CCL2 contributes to inflammatory pain via modulating Nav channel activity of primary afferent neurons. Chronic inflammatory pain was induced in rats by intraplantar injection of the complete Freud adjuvant (CFA) to one of the hind paws. Control rats received intraplantar injection of equal volume of saline. A significant increase of CCL2 mRNA and CCL2 receptor (CCR2) protein expression was detected in the ipsilateral dorsal root ganglion (DRG) in CFA-treated rats. Intraplantar injection of CCL2 protein in the control rats had minimal effect on the paw withdrawal threshold (PWT) in response to mechanical stimulation. However, in CFA-treated rats, intraplantar CCL2 led to an increase in pain responses. Patch-clamp recording of acutely dissociated DRG neurons revealed that CCL2 had minimum effect on the excitability of sensory neurons from control rats. However, CCL2 directly depolarized a large proportion of small to medium-sized sensory neurons from CFA-treated rats. In addition, CCL2 was found to enhance whole-cell TTX-sensitive sodium currents without significantly affecting the TTX-resistant sodium currents and the potassium currents. These results are in agreement with previous reports concerning the involvement of CCL2-CCR2 signaling in inflammatory hyperalgesia and further indicate that enhanced TTX-sensitive channel activity may partly underlie the pronociceptive effects of CCL2.


Asunto(s)
Quimiocina CCL2/farmacología , Inflamación/metabolismo , Neuronas Aferentes/efectos de los fármacos , Dolor/metabolismo , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Sinergismo Farmacológico , Adyuvante de Freund , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas Aferentes/metabolismo , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/genética
20.
Int J Med Sci ; 12(7): 559-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26180512

RESUMEN

BACKGROUND: Propofol may result in hypotension and respiratory depression, while etomidate is considered to be a safe induction agent for haemodynamically unstable patients because of its low risk of hypotension. We hypothesized that etomidate anesthesia during ERCP caused more stable haemodynamic responses compared with propofol. The primary endpoint was to compare the haemodynamic effects of etomidate vs. propofol in ERCP cases. The secondary endpoint was overall survival. METHODS: A total of 80 patients undergoing ERCP were randomly assigned to an etomidate or propofol group. Patients in the etomidate group received etomidate induction and maintenance during ERCP, and patients in the propofol group received propofol induction and maintenance. Cardiovascular parameters and procedure-related time were measured and recorded during ERCP. RESULTS: The average percent change to baseline in MBP was -8.4±7.8 and -14.4±9.4 with P = 0.002, and in HR was 1.8±16.6 and 2.4±16.3 with P = 0.874 in the etomidate group and the propofol group, respectively. MBP values in the etomidate group decreased significantly less than those in the propofol group (P<0.05). The ERCP duration and recovery time in both groups was similar. There was no significant difference in the survival rates between groups ( p = 0.942). CONCLUSIONS: Etomidate anesthesia during ERCP caused more stable haemodynamic responses compared with propofol.


Asunto(s)
Anestesia/efectos adversos , Etomidato/administración & dosificación , Hemodinámica/efectos de los fármacos , Propofol/administración & dosificación , Adolescente , Adulto , Anciano , Presión Sanguínea/efectos de los fármacos , Colangiopancreatografia Retrógrada Endoscópica , Etomidato/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Propofol/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA