Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Anesthesiology ; 140(6): 1134-1152, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38498811

RESUMEN

BACKGROUND: Dexmedetomidine has repeatedly shown to improve anxiety, but the precise neural mechanisms underlying this effect remain incompletely understood. This study aims to explore the role of corticotropin-releasing hormone-producing hypothalamic paraventricular nucleus (CRHPVN) neurons in mediating the anxiolytic effects of dexmedetomidine. METHODS: A social defeat stress mouse model was used to evaluate the anxiolytic effects induced by dexmedetomidine through the elevated plus maze, open-field test, and measurement of serum stress hormone levels. In vivo Ca2+ signal fiber photometry and ex vivo patch-clamp recordings were used to determine the excitability of CRHPVN neurons and investigate the specific mechanism involved. CRHPVN neuron modulation was achieved through chemogenetic activation or inhibition. RESULTS: Compared with saline, dexmedetomidine (40 µg/kg) alleviated anxiety-like behaviors. Additionally, dexmedetomidine reduced CRHPVN neuronal excitability. Chemogenetic activation of CRHPVN neurons decreased the time spent in the open arms of the elevated plus maze and in the central area of the open-field test. Conversely, chemogenetic inhibition of CRHPVN neurons had the opposite effect. Moreover, the suppressive impact of dexmedetomidine on CRHPVN neurons was attenuated by the α2-receptor antagonist yohimbine. CONCLUSIONS: The results indicate that the anxiety-like effects of dexmedetomidine are mediated via α2-adrenergic receptor-triggered inhibition of CRHPVN neuronal excitability in the hypothalamus.


Asunto(s)
Ansiedad , Dexmedetomidina , Neuronas , Núcleo Hipotalámico Paraventricular , Estrés Psicológico , Animales , Masculino , Ratones , Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Hormona Liberadora de Corticotropina/metabolismo , Dexmedetomidina/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Estrés Psicológico/psicología
2.
Biochem Biophys Res Commun ; 643: 147-156, 2023 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-36609155

RESUMEN

An increasing number of experimental and clinical observation suggest that the use of anaesthetics is closely associated with postoperative central nervous system (CNS) complications, such as delirium and cognitive dysfunction. Brain energy rescue is an emerging therapeutic strategy for central nervous system disease (CNSDs). However, the effect of anaesthetics on nerve cell energy utilisation, especially microglia, and its potential effects on cell function still unclear. Elucidating the effects of anaesthetics on lipid droplets, which are specific lipid storage organs, and phagocytosis of microglia is crucial to discover a new therapeutic concept for postoperative CNS complications. Here, we studied the effects of the commonly used anaesthetic midazolam on lipid droplets and phagocytosis in immortalised microglial BV2 cells. Lipid droplets were assessed by flow cytometry and triglyceride quantification. The phagocytosis of BV2 cells was evaluated by detecting their phagocytosis by latex beads. Additionally, the autophagy of BV2 cells was evaluated by western blot and observation under microscopy. Our results showed that midazolam caused lipid droplet accumulation and reduced phagocytosis in BV2 cells, and inhibition of lipid droplet accumulation partially restored phagocytosis. Furthermore, midazolam blocks autophagic degradation by increasing phosphorylated TFEB in BV2 cells, inhibition of midazolam-increased phosphorylated TFEB might contribute to the improvement of autophagic flux by rapamycin. Moreover, promoting autophagy reverse the lipid droplet accumulation and phagocytosis decrease. This study suggests autophagy is a target for attenuating lipid droplet accumulation, normal degradation of lipid droplets is important for maintaining microglia phagocytosis and attenuating the side effects of midazolam on the CNS.


Asunto(s)
Gotas Lipídicas , Midazolam , Midazolam/farmacología , Fagocitosis , Autofagia , Microglía/metabolismo
3.
J Nanobiotechnology ; 21(1): 50, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765361

RESUMEN

Postoperative pain (POP) can promote tumor recurrence and reduce the cancer patient's quality of life. However, POP management has always been separated from tumor treatment in clinical practice, and traditional postoperative analgesia using opioids is still unsatisfactory for patients, which is not conducive to tumor treatment. Here, ropivacaine, a popular amide-type LA, was introduced into a Pluronic F127 hydrogel. Postoperative analgesia with ropivacaine-loaded hydrogels reduced the incidence of high-dose ropivacaine-induced convulsions and prolonged pain relief for more than 16 h. More interestingly, ropivacaine-loaded hydrogel was found to upregulate major histocompatibility complex class I (MHC-I) in tumor cells by impairing autophagy. Therefore, a hydrogel co-dopped with ropivacaine and TLR7 agonist imiquimod (PFRM) was rationally synthesized. After postoperative analgesia with PFRM, imiquimod primes tumor-specific CD8+T cells through promoting DCs maturation, and ropivacaine facilitates tumor cells recognition by primed CD8+T cells through upregulating MHC-I. Consequently, postoperative analgesia with PFRM maximumly increases CD8+T cells infiltration into residual tumor tissue and prevents tumor recurrence. Overall, this study for the first time provides an LA-based approach for simultaneous long-lasting postoperative analgesia and prevention of tumor recurrence.


Asunto(s)
Analgesia , Anestésicos Locales , Humanos , Anestésicos Locales/farmacología , Anestésicos Locales/uso terapéutico , Ropivacaína/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/prevención & control , Imiquimod , Calidad de Vida , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Linfocitos T CD8-positivos , Linfocitos T
4.
J Nanobiotechnology ; 21(1): 462, 2023 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-38041074

RESUMEN

Chemotherapy can cause severe pain for patients, but there are currently no satisfactory methods of pain relief. Enhancing the efficacy of chemotherapy to reduce the side effects of high-dose chemotherapeutic drugs remains a major challenge. Moreover, the treatment of chemotherapy-induced peripheral neuropathic pain (CIPNP) is separate from chemotherapy in the clinical setting, causing inconvenience to cancer patients. In view of the many obstacles mentioned above, we developed a strategy to incorporate local anesthetic (LA) into a cisplatin-loaded PF127 hydrogel for painless potentiated chemotherapy. We found that multiple administrations of cisplatin-loaded PF127 hydrogels (PFC) evoked severe CIPNP, which correlated with increased pERK-positive neurons in the dorsal root ganglion (DRG). However, incorporating ropivacaine into the PFC relieved PFC-induced CIPNP for more than ten hours and decreased the number of pERK-positive neurons in the DRG. Moreover, incorporating ropivacaine into the PFC for chemotherapy is found to upregulate major histocompatibility complex class I (MHC-I) expression in tumor cells and promote the infiltration of cytotoxic T lymphocytes (CD8+ T cells) in tumors, thereby potentiating chemotherapy efficacy. This study proposes that LA can be used as an immunemodulator to enhance the effectiveness of chemotherapy, providing new ideas for painless cancer treatment.


Asunto(s)
Antineoplásicos , Neuralgia , Humanos , Ropivacaína/efectos adversos , Cisplatino , Linfocitos T CD8-positivos/metabolismo , Hidrogeles , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Antineoplásicos/efectos adversos
5.
Clin Exp Pharmacol Physiol ; 50(8): 664-676, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37254848

RESUMEN

Ischemia-reperfusion (I/R) injury is a common pathophysiological condition in ischemic stroke, involving various pathophysiological events, such as inflammation, cytotoxicity, neuronal loss and disruption of the blood-brain barrier (BBB). Rosavin is the major bioactive ingredient of Rhodiola Rosea L. with multiple therapeutic effects. The purpose of this was to investigate the role of rosavin in I/R-induced cerebral injury. A cell oxygen-glucose deprivation and reoxygenation (OGD/R) model and a mouse middle cerebral artery occlusion (MCAO) model were established to induce I/R injury in vitro and in vivo, respectively. MCAO-treated mice and OGD/R-challenged human brain microvascular endothelial cells (HBMVECs) were administrated with or without rosavin at various concentrations. Rosavin-treated mice showed reduced infarct volume, neuronal loss and neuronal cytotoxicity in I/R-injured brains. Rosavin treatment downregulated the expression of pro-inflammatory cytokines, reduced apoptosis and inhibited the activation of nuclear factor κ B in I/R-injured mice and HBMVECs. Administration with rosavin also alleviated mouse brain oedema and upregulated tight junction proteins in mouse brains after I/R injury, suggesting that rosavin protected mice against I/R-induced BBB disruption. Further analysis revealed that rosavin reduced the BBB permeability in I/R-injured mice and HBMVECs by inhibiting autophagy. Moreover, rosavin intervention inhibited I/R injury-induced activation of the mitogen-activated protein kinase (MAPK) pathway and upregulation of matrix metalloproteinases in both mouse and cell models. In conclusion, rosavin protects the BBB against I/R injury possibly by regulating the MAPK pathway. The above results provide a rationale for further exploration of rosavin as a therapeutic candidate for cerebral I/R injury.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Ratones , Humanos , Animales , Barrera Hematoencefálica/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Endoteliales/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Reperfusión , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Metaloproteinasas de la Matriz
6.
Nanotechnology ; 32(26)2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33472187

RESUMEN

The aim of this study is to evaluate the effect of rare earth upconversion nanoparticles (UCNs) on hepatic ischemia reperfusion injury (IRI) and explore its possible mechanism. Hepatic IRI seriously affects the prognosis of patients undergoing liver surgery. Liver-resident Kupffer cells have been reported to promote IRI. Nanomedicines are known to be effective in the treatment of liver diseases, however, Kupffer cell-targeting nanomedicines for the treatment of IRI are yet to be developed. As potential bioimaging nanomaterials, UCNs have been found to specifically deplete Kupffer cells, but the underlying mechanism is unknown. In this study, we found that UCNs specifically depleted Kupffer cells by pyroptosis, while the co-administration of the caspase-1 inhibitor VX-765 rescued the UCN-induced Kupffer cell pyroptosis in mice. Furthermore, the pre-depletion of Kupffer cells by the UCNs significantly suppressed the release of inflammatory cytokines and effectively improved hepatic IRI. The rescue of the pyroptosis of the Kupffer cells by VX-765 abrogated the protective effect of UCNs on the liver. These results suggest that UCNs are highly promising for the development of Kupffer cell-targeting nanomedicines for intraoperative liver protection.


Asunto(s)
Macrófagos del Hígado/efectos de los fármacos , Hígado/efectos de los fármacos , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Daño por Reperfusión/terapia , Animales , Macrófagos del Hígado/patología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Piroptosis/efectos de los fármacos , Daño por Reperfusión/patología
7.
Biochem Biophys Res Commun ; 514(3): 953-959, 2019 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-31092327

RESUMEN

Remifentanil postconditioning (RPC) has been shown to provide potent cardioprotection against ischemia/reperfusion (I/R) injury, but the underlying mechanism has not been fully elucidated. The current study was designed to investigate whether RPC protects cardiomyocytes against I/R injury through enhancement of autophagic flux. H9c2 cardiomyocytes were exposed to hypoxia/reoxygenation (H/R) to mimic myocardial I/R injury in vitro. Autophagosome formation was evaluated by detecting of light chain 3 (LC3) puncta number and LC3Ⅱ levels using immunofluorescence and western blotting, respectively. Additionally, dual fluorescent staining of LC3 and lysosomal-associated membrane protein 2, a lysosomal marker protein, were used to detect autolysosome formation. Moreover, autophagic flux integrity was tracked using changes in LC3Ⅱ and p62 levels. Lastly, myocardial injury was detected by Hoechst 33342 and propidium iodide staining and MTT assay. The results showed that RPC increased autophagosome formation and promoted autophagosome-lysosome fusion, thereby improving autophagic flux in H9c2 cells. Reversal of these effect by bafilomycin A1 or chloroquine co-administration at reoxygenation onset indicated that RPC improved the impaired autophagic flux following H/R injury. Induction of autophagy was associated with increased cell viability and decreased apoptosis. Autophagy inhibition with bafilomycin A1 or chloroquine and ATG7shRNA significantly abolished RPC-induced cardioprotection. In conclusion, our finding that RPC can protect cardiomyocytes against H/R injury through enhancement of autophagic flux suggests a new mechanism for myocardial protection of opioid postconditioning.


Asunto(s)
Autofagia/efectos de los fármacos , Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Remifentanilo/farmacología , Analgésicos Opioides/farmacología , Animales , Línea Celular , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Ratas
8.
Acta Pharmacol Sin ; 40(6): 755-761, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30367153

RESUMEN

Propofol is an intravenous anesthetic that can active γ-aminobutyric acid A (GABAA) receptors and generate sedative-hypnotic effects. Propofol has been widely applied clinically to achieve sedation comparable to sleep in humans. The basal forebrain (BF) is a brain region that plays an important role in sleep-wake regulation. Previous studies suggest that propofol affects the sleep-wake circuit via the BF; however, the mechanism remains elusive. In the current study we investigated the effects of propofol on the inherent properties of cholinergic neurons and their ability to convert excitatory inputs into spikes in mouse BF slices using whole-cell patch clamp recordings. Bath application of propofol (10 µM) significantly elevated the threshold potentials (Vts), decreased the number of spikes in response to a depolarizing current injection, and augmented the inter-spike intervals (ISIs), energy barrier (Vts-Vrs), and absolute refractory periods (ARPs). These effects were eliminated by co-application of a GABAA receptor antagonist picrotoxin (50 µM). Altogether, our results reveal that propofol decreases the excitability of cholinergic neurons in mouse BF via GABAA receptors.


Asunto(s)
Anestésicos Intravenosos/farmacología , Prosencéfalo Basal/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Propofol/farmacología , Receptores de GABA-A/metabolismo , Animales , Ratones Endogámicos C57BL
9.
Cell Physiol Biochem ; 48(2): 683-691, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30025395

RESUMEN

BACKGROUND/AIMS: Autophagy is a well-known pathway to "clean" the misfolded mutant huntingtin protein (mHtt), which plays a considerable role in polyglutamine diseases. To date, there have been few studies of the choice of anesthetic during surgery in patients with polyglutamine diseases and evaluation of the effects and underlying mechanisms of anesthetics in these patients. METHODS: GFP-Htt (Q74)-PC12 cells, which stably express green fluorescent protein-tagged Htt protein containing 74 glutamine repeating units, were used throughout this study. Cells were treated with 15 µM midazolam and 100 mM trehalose (positive control), and the induction of autophagy and autophagic degradation were assessed by detecting changes in autophagy-related proteins and substrates, and cell viability was assessed using the MTT assay. Overexpression of cathepsin D by plasmid transfection was used to restore midazolam-impaired autophagic degradation. RESULTS: Midazolam increased intracellular mHtt levels in a time- and dose-dependent manner. Additionally, enhancing or blocking autophagic flux by trehalose or chloroquine could decrease or increase midazolam-induced mHtt elevation, respectively. Midazolam induced autophagy in the mTOR-dependent signaling pathway, but autophagic degradation was impaired, with a continuous rise in p62 and LC3 II levels and decrease in cathepsin D. However, overexpression of cathepsin D reversed the effects of midazolam. Midazolam led to a 20% decrease in GFP-Htt (Q74)-PC12 cell viability, which could be abrogated by overexpression of cathepsin D. CONCLUSIONS: Midazolam increased mHtt levels and decreased Htt (Q74)-PC12 cell viability via impairment of autophagic degradation, which could be restored by overexpression of cathepsin D.


Asunto(s)
Autofagia/efectos de los fármacos , Proteína Huntingtina/metabolismo , Midazolam/farmacología , Animales , Catepsina D/genética , Catepsina D/metabolismo , Supervivencia Celular/efectos de los fármacos , Proteína Huntingtina/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Células PC12 , Proteolisis/efectos de los fármacos , Ratas , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
10.
Zool Res ; 45(3): 679-690, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38766749

RESUMEN

General anesthesia is widely applied in clinical practice. However, the precise mechanism of loss of consciousness induced by general anesthetics remains unknown. Here, we measured the dynamics of five neurotransmitters, including γ-aminobutyric acid, glutamate, norepinephrine, acetylcholine, and dopamine, in the medial prefrontal cortex and primary visual cortex of C57BL/6 mice through in vivo fiber photometry and genetically encoded neurotransmitter sensors under anesthesia to reveal the mechanism of general anesthesia from a neurotransmitter perspective. Results revealed that the concentrations of γ-aminobutyric acid, glutamate, norepinephrine, and acetylcholine increased in the cortex during propofol-induced loss of consciousness. Dopamine levels did not change following the hypnotic dose of propofol but increased significantly following surgical doses of propofol anesthesia. Notably, the concentrations of the five neurotransmitters generally decreased during sevoflurane-induced loss of consciousness. Furthermore, the neurotransmitter dynamic networks were not synchronized in the non-anesthesia groups but were highly synchronized in the anesthetic groups. These findings suggest that neurotransmitter dynamic network synchronization may cause anesthetic-induced loss of consciousness.


Asunto(s)
Anestésicos por Inhalación , Ratones Endogámicos C57BL , Neurotransmisores , Propofol , Sevoflurano , Sevoflurano/farmacología , Animales , Propofol/farmacología , Neurotransmisores/metabolismo , Ratones , Anestésicos por Inhalación/farmacología , Anestésicos Intravenosos/farmacología , Masculino , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
11.
Cerebellum ; 12(6): 862-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23775547

RESUMEN

The spike propagation on nerve axons, like synaptic transmission, is essential to ensure neuronal communication. The secure propagation of sequential spikes toward axonal terminals has been challenged in the neurons with a high firing rate, such as cerebellar Purkinje cells. The shortfall of spike propagation makes some digital spikes disappearing at axonal terminals, such that the elucidation of the mechanisms underlying spike propagation reliability is crucial to find the strategy of preventing loss of neuronal codes. As the spike propagation failure is influenced by the membrane potentials, this process is likely caused by altering the functional status of voltage-gated sodium channels (VGSC). We examined this hypothesis in Purkinje cells by using pair-recordings at their somata and axonal blebs in cerebellar slices. The reliability of spike propagation was deteriorated by elevating spike frequency. The frequency-dependent reliability of spike propagation was attenuated by inactivating VGSCs and improved by removing their inactivation. Thus, the functional status of axonal VGSCs influences the reliability of spike propagation.


Asunto(s)
Potenciales de Acción/fisiología , Axones/fisiología , Cerebelo/citología , Células de Purkinje/citología , Canales de Sodio Activados por Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Biofisica , Biotina/análogos & derivados , Biotina/metabolismo , Venenos de Cnidarios/farmacología , Estimulación Eléctrica , Técnicas In Vitro , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Bloqueadores de los Canales de Sodio/farmacología
12.
Mitochondrial DNA B Resour ; 8(6): 686-690, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37359090

RESUMEN

Lithocarpus konishii, a rare species endemic to islands in South China, was evaluated as a vulnerable species (VU) by the 'China Species Red List.' Here, we first presented the complete chloroplast genome sequence of L. konishii. The chloroplast genome was 161,059 bp in length with 36.76% GC content, containing a small single-copy region (SSC, 18,967 bp), a large single-copy region (LSC, 90,250 bp), and a pair of inverted repeats (IRs, 25,921 bp each). A total of 139 genes were predicted, including 87 protein-coding genes (CDS), 8 rRNAs, and 44 tRNAs. Based on the concatenated shared unique CDS sequence dataset, maximum-likelihood and Bayesian inference methods were used to build the phylogenetic trees of 18 species from the Fagaceae family. Results indicated that L. konishii is closely related to L. longnux and L. pachyphyllus var. fruticosus, and forms a monophyly of the subfamily Castaneoideae with Castanopsis and Castanea. This study provides a theoretical basis for the conservation genomics of this endangered plant.

13.
iScience ; 26(4): 106414, 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37034991

RESUMEN

Chemotherapy-induced peripheral neuropathic pain (CIPNP) is a progressive and disturbing peripheral neuropathy with currently no effective treatment. Aberrant production of reactive oxygen species (ROS) in macrophages near peripheral nerves plays a dominant role in CIPNP; however, traditional ROS scavengers have difficulty maintaining viability to target macrophages in vivo. Mannose-coated superparamagnetic iron oxide nanoparticles (mSPIONs) were synthesized to treat CIPNP. The anti-ROS and anti-inflammatory effects of mSPIONs were assessed in J774A.1 cells and sciatic nerves in a nociception mouse model induced with vincristine (VCR). We found that the mSPIONs significantly reduced ROS levels in vitro and in vivo. Furthermore, mSPIONs administration specifically reduced IL-6 and TNF-α levels in macrophages near the sciatic nerve and relieved VCR-induced peripheral neuropathic pain. Inhibition of the VCR-upregulated HIF1α/NF-κB signaling pathway may be involved in the alleviation of inflammation. These results provide a new approach for relieving CIPNP using a nanozyme.

14.
Mater Today Bio ; 19: 100568, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36846307

RESUMEN

Postoperative cognitive dysfunction (POCD) is associated with increased postoperative morbidity and mortality in patients. Excessive production of reactive oxygen species (ROS) and the consequent inflammatory response in the postoperative brain play crucial roles in the development of POCD. However, effective ways to prevent POCD have yet to be developed. Moreover, effective penetration of the blood-brain barrier (BBB) and maintaining viability in vivo are major challenges for preventing POCD using traditional ROS scavengers. Herein, mannose-coated superparamagnetic iron oxide nanoparticles (mSPIONs) were synthesized by co-precipitation method. The BBB penetration of mSPIONs was verified through fluorescent imaging and ICP-MS quantification. The ROS scavenging and anti-inflammatory of mSPIONs were evaluated in H2O2-treated J774A.1 â€‹cells and in tibial fracture mice model. The novel object recognition (NOR) and trace-fear conditioning (TFC) were used to test the cognitive function of postoperative mice. The average diameter of mSPIONs was approximately 11 â€‹nm. mSPIONs significantly reduced ROS levels in H2O2-treated cells and in hippocampus of surgical mice. mSPIONs administration reduced the levels of IL-1ß and TNF-α in the hippocampus and inhibited surgery-upregulated HIF1-α/NF-κB signaling pathway. Moreover, mSPIONs significantly improved the cognitive function of postoperative mice. This study provides a new approach for preventing POCD using a nanozyme.

15.
Theranostics ; 13(7): 2226-2240, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37153743

RESUMEN

Rationale: Tumor ablation can cause severe pain to patients, but there is no satisfactory means of analgesia available. In addition, recurrence of residual tumors due to incomplete ablation threatens patient safety. Photothermal therapy (PTT), a promising approach for tumor ablation, also faces the aforementioned problems. Therefore, developing novel photothermal agents that can efficiently relieve PTT-associated pain and potentiate the PTT efficacy are urgently needed. Methods: The Pluronic F127 hydrogel doped with indocyanine green (ICG) was served as photothermal agent for PTT. Mouse model that inoculation of tumor near the sciatic nerve was constructed to assess the PTT-evoked pain. Subcutaneous and sciatic nerve vicinal tumor-bearing mice were used to test the efficacy of PTT. Results: PTT-evoked pain depends on an increase in tumor temperature and is accompanied by the activation of TRPV1. A simple introduction of local anesthetic (LA) ropivacaine into ICG-loaded hydrogels relieves PTT-induced pain and exerts long-lasting analgesia compared with opioid analgesia. More interestingly, ropivacaine upregulates major histocompatibility complex class I (MHC-I) in tumor cells by impairing autophagy. Therefore, a hydrogel co-doped with ropivacaine, TLR7 agonist imiquimod and ICG was rationally designed. In the hydrogel system, imiquimod primes tumor-specific CD8+ T cells through promoting DCs maturation, and ropivacaine facilitates tumor cells recognition by primed CD8+ T cells through upregulating MHC-I. Consequently, the hydrogel maximumly increases CD8+ T cells infiltration into tumor and potentiates PTT efficacy. Conclusion: This study for the first time provides an LA-dopped photothermal agents for painless PTT and innovatively proposes that a LA can be used as an immunomodulator to potentiate the PTT efficacy.


Asunto(s)
Neoplasias , Fototerapia , Animales , Ratones , Hidrogeles , Terapia Fototérmica , Ropivacaína , Linfocitos T CD8-positivos , Imiquimod , Neoplasias/terapia , Verde de Indocianina/uso terapéutico , Analgésicos , Dolor
16.
Front Neurosci ; 16: 828278, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573301

RESUMEN

Inflammatory pain is the most common type of pain encountered in clinical practice; however, the currently available treatments are limited by insufficient efficacy and side effects. Therefore, new methods to relieve inflammatory pain targeting new mechanisms are urgently needed. Preclinical investigations have shown that CR (calorie restriction) exerts analgesic effects in neuropathic and cancer pain; however, the effect of CR on chronic inflammatory pain remains unknown. During calorie restriction, autophagy, a lysosome-dependent degradation process, can be activated to support cell survival. In the present study, we investigated the analgesic effects of CR on complete Freund's adjuvant (CFA)-induced inflammatory pain. The accumulation of LC3-II and p62 showed impaired autophagic flux in the ipsilateral spinal cord of mice with CFA-induced inflammatory pain. CR alleviated mechanical allodynia and thermal hyperalgesia and reduced paw edema and pro-inflammatory factors following CFA administration. CR exerted an analgesic effect by restoring autophagic flux in the spinal cord. Regarding the mechanisms underlying the analgesic effects of CR, ß-hydroxybutyric acid (BHB) was studied. CR increased BHB levels in the ipsilateral spinal cord. Furthermore, exogenous BHB administration exerted an analgesic effect by restoring autophagic flux in the spinal cords of CFA-induced inflammatory pain mice. Taken together, these results illustrated that CR relieved inflammatory pain by restoring autophagic flux in the spinal cord, while BHB controlled the benefits of CR, suggesting that CR or BHB might be a promising treatment for inflammatory pain.

17.
ACS Chem Neurosci ; 13(23): 3534-3543, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36441865

RESUMEN

The wide disparity in outcomes of Alzheimer's disease (AD) treatment from preclinical to clinical studies suggests an urgent need for more effective therapeutic targets and approaches to treat AD. CaMKII is a potential target for AD therapy; however, conflicting reports on the relationship between CaMKII and AD suggest a lack of deeper understanding of the interaction between CaMKII and AD. In addition to the lack of effective therapeutic targets, pharmacokinetic limitations of neuroprotective drugs, such as low lipophilicity to cross blood brain barrier, need to be urgently addressed in the practice of AD therapy. In this study, we prepared a carbon-based nanoparticle, Nano C60, and demonstrated that Nano C60 treatment promoted the translocation of phosphorylated CaMKIIα from the cytoplasm to the synapse in Aß42 oligomers-treated cells and APP/PS1 mice. As a result, Nano C60 administration significantly improved spatial learning and memory in APP/PS1 mice. Our study suggests that synaptic-activated CaMKII may be more important than total CaMKII in AD treatment and provides a new strategy for AD therapy.


Asunto(s)
Cognición , Ratones , Animales , Ratones Transgénicos
18.
Theranostics ; 10(11): 4885-4899, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32308756

RESUMEN

Autophagy allows cancer cells to respond changes in nutrient status by degrading and recycling non-essential intracellular contents. Inhibition of autophagy combined with nutrient deprivation is an effective strategy to treat cancer. Pain is a primary determinant of poor quality of life in advanced cancer patients, but there is currently no satisfactory treatment. In addition, effective treatment of cancer does not efficiently relieve cancer pain, but may increase pain in many cases. Hence, few studies focus on simultaneous cancer therapy and pain relief, and made this situation even worse. Method: Ropivacaine was loaded into tumor-active targeted liposomes. The cytotoxicity of ropivacaine-based combination therapy in B16 and HeLa cells were tested. Moreover, a mice model of cancer pain which was induced by inoculation of melanoma near the sciatic nerve was constructed to assess the cancer suppression and pain relief effects of ropivacaine-based combination therapy. Results: Ropivacaine and ropivacaine-loaded liposomes (Rop-DPRL) were novelly found to damage autophagic degradation. Replicated administration of Rop-DPRL and calorie restriction (CR) could efficiently repress the development of tumor. In addition, administration of Rop-DPRL could relieve cancer pain with its own analgestic ability in a short duration, while repeated administration of Rop-DPRL and CR resulted in continuous alleviation of cancer pain through reduction of VEGF-A levels in advanced cancer mice. Further, dual inhibition of phosphorylation of STAT3 at Tyr705 and Ser727 by Rop-DPRL and CR contribute to the reduction of VEGF-A. Conclusion: Combination therapy with Rop-DPRL and nutrient deprivation simultaneously suppresses cancer growth and relieves cancer pain.


Asunto(s)
Autofagia , Restricción Calórica , Dolor en Cáncer/terapia , Liposomas/administración & dosificación , Melanoma/terapia , Ropivacaína/farmacología , Nervio Ciático/patología , Neoplasias del Cuello Uterino/terapia , Anestésicos Locales/farmacología , Animales , Dolor en Cáncer/etiología , Dolor en Cáncer/patología , Línea Celular Tumoral , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Liposomas/química , Masculino , Melanoma/complicaciones , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias del Cuello Uterino/complicaciones , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Theranostics ; 10(26): 11908-11920, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33204319

RESUMEN

Rationale: Graphene oxide (GO) based nanomaterials have shown potential for the diagnosis and treatment of amyloid-ß (Aß)-related diseases, mainly on Alzheimer's disease (AD). However, these nanomaterials have limitations. How GO is beneficial to eliminate Aß burden, and its physiological function in Aß-related diseases, still needs to be investigated. Moreover, postoperative cognitive dysfunction (POCD) is an Aß-related common central nervous system complication, however, nanomedicine treatment is lacking. Methods: To evaluate the effects of GO on Aß levels, HEK293T-APP-GFP and SHSY5Y-APP-GFP cells are established. Intramedullary fixation surgery for tibial fractures under inhalation anesthesia is used to induce dysfunction of fear memory in mice. The fear memory of mice is assessed by fear conditioning test. Results: GO treatment maximally alleviated Aß levels by simultaneously reducing Aß generation and enhancing its degradation through inhibiting ß-cleavage of amyloid precursor protein (APP) and improving endosomal Aß delivery to lysosomes, respectively. In postoperative mice, the hippocampal Aß levels were significantly increased and hippocampal-dependent fear memory was impaired. However, GO administration significantly reduced hippocampal Aß levels and improved the cognitive function of the postoperative mice. Conclusion: GO improves fear memory of postoperative mice by maximally alleviating Aß accumulation, providing new evidence for the application of GO-based nanomedicines in Aß-related diseases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Fijación Intramedular de Fracturas/efectos adversos , Grafito/administración & dosificación , Nanopartículas/administración & dosificación , Complicaciones Cognitivas Postoperatorias/tratamiento farmacológico , Péptidos beta-Amiloides/análisis , Animales , Modelos Animales de Enfermedad , Miedo , Femenino , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Memoria/efectos de los fármacos , Ratones , Complicaciones Cognitivas Postoperatorias/etiología , Complicaciones Cognitivas Postoperatorias/patología , Fracturas de la Tibia/cirugía
20.
Biosci Rep ; 39(11)2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31710085

RESUMEN

Sevoflurane was found to show protective roles in mice with asthma, however, the mechanism of which needs further exploring. Aquaporins (AQPs) have been demonstrated to be involved in the pathogenesis of asthma, while endoplasmic reticulum stress has been reported to be related to many inflammatory diseases and involved in protein processing, including AQPs. The present study aimed to determine the role of sevoflurane in AQPs (AQP1,3,4,5) expression in mice with allergic airway inflammation and the probable mechanism. The increased number of inflammatory cells infiltrating the lung tissue, and the elevated levels of tumor necrosis factor-α (TNF-α) and interleukin (IL) 13 (IL-13) were all decreased after sevoflurane treatment (all P<0.05). Meanwhile, mRNA levels of AQP1 and AQP5 but not AQP3 and AQP4 were decreased in ovalbumin (OVA)-induced allergic mice lung. Both the decreased mRNA expression and protein levels of AQP1 and AQP5 in allergic lung tissues were reversed by sevoflurane treatment. Furthermore, we established that sevoflurane inhibited the OVA-induced protein increase in the endoplasmic reticulum (ER) stress markers BiP and C/EBP homologous protein (CHOP). Collectively, these findings suggested that sevoflurane modulated the expression and protein level of AOPs (AQP1, AQP5) as well as inhibited ER stress response in OVA-induced allergic airway inflammation of mice.


Asunto(s)
Acuaporinas/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hipersensibilidad/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Sevoflurano/farmacología , Animales , Asma/tratamiento farmacológico , Asma/metabolismo , Citocinas/metabolismo , Femenino , Hipersensibilidad/metabolismo , Inflamación/metabolismo , Interleucina-13/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción CHOP/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA