Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Clin Nurs ; 32(7-8): 1014-1024, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35229381

RESUMEN

AIMS AND OBJECTIVES: This systematic review and meta-analysis aimed to compare the incidence of PVC-related complications between catheterisation in the forearm and back of the hand in adult patients. BACKGROUND: A peripheral intravenous catheter (PVC) is often inserted as part of care during patients' hospitalisation. The catheter is typically inserted in the forearm or at the back of the hand in usual practice. Studies have not yet reached a consensus on the optimal insertion site in any clinical setting. DESIGN: We performed a systematic review and meta-analysis based on PRISMA guidelines. METHODS: We searched the following electronic databases: PubMed, Cochrane Library, Embase, and CINAHL. Randomised controlled trials, cohort studies, case-control studies and cross-sectional studies from inception to July 2021 reporting the incidence of PVC-related complications at the forearm and back of the hand were included. Fixed-effects models and random-effects models were used to derive the pooled risk ratios. RESULTS: Twenty-four studies involving 16562 PVCs met our inclusion criteria. The meta-analysis showed that compared with PVC placement in the back of the hand, placement in the forearm was associated with a higher incidence of total complications and infiltration/extravasation. However, the differences between the PVC indwelling sites were not significant (total complications: P = 0.43; phlebitis: P = 0.35; infiltration/extravasation: P = 0.51). Both incidence of total complications and infiltration/extravasation analyses showed high heterogeneity (total complications: I2  = 60%; infiltration/extravasation: I2  = 58%). CONCLUSION: Available evidence suggests that there is no significant difference between PVC placement in the forearm and at the back of the hand in terms of the incidence of complications, thus making both approaches suitable. RELEVANCE TO CLINICAL PRACTICE: For patients who need indwelling PVC, medical staff can choose the best indwelling site, and both forearm and back of the hand are suitable.


Asunto(s)
Catéteres , Hospitalización , Humanos , Adulto , Estudios Transversales , Estudios de Casos y Controles , Consenso
2.
Mol Biol Rep ; 49(8): 7697-7707, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35655056

RESUMEN

BACKGROUND: There is a relationship between autophagy and the occurrence, maintenance, and progression of several neurodegenerative diseases. The activation of microglia after ischemia contributes to neuronal injury via proinflammatory cytokines and neurotoxic elements. The purpose of this study was to evaluate the function of autophagy in the microglia-mediated death of neuronal cells. METHODS AND RESULTS: Microglial activation by oxygen/glucose deprivation induced both apoptosis and autophagy in neuron-like PC12 cells. Microglia-derived interleukin (IL)-6 induced PC12 cell apoptosis in vitro; however, this effect was inhibited by the autophagy inhibitor chloroquine. Further analysis demonstrated that miR-30d in PC12 cells suppressed microglia-induced PC12 apoptosis and autophagy by directly targeting autophagy protein 5. Moreover, microglia-derived IL-6 activated signal transducer and activator of transcription 3 (STAT3), which can then directly repress miR-30d genes via a conserved STAT3-binding site in its promoter, thereby promoting PC12 cell autophagy and apoptosis. CONCLUSIONS: Our study identified IL-6-dependent autophagy-related signaling between microglia and neurons, which contributed to neuronal apoptosis. Importantly, we also provided potential therapeutic targets for ischemic treatment via the interruption of proinflammatory signaling.


Asunto(s)
MicroARNs , Microglía , Animales , Apoptosis , Hipoxia/metabolismo , Interleucina-6/metabolismo , Isquemia , MicroARNs/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Ratas , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
3.
J Neurochem ; 132(6): 713-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25545945

RESUMEN

Cerebral hypoxia/ischemia rapidly induces inflammation in the brain, which is characterized by microglial activation and the release of inflammatory cytokines. We have previously demonstrated that miR-181c can directly regulate tumor necrosis factor (TNF)-α production post-transcriptionally. Here, we determined that hypoxia up-regulated TLR4 expression but down-regulated miR-181c expression in primary microglia. We also demonstrated that miR-181c suppresses TLR4 by directly binding its 3'-untranslated region. In addition, miR-181c inhibited NF-κB activation and the downstream production of proinflammatory mediators, such as TNF-α, IL-1ß, and iNOS. Knocking down TLR4 in microglia significantly decreased TLR4 expression and inhibited NF-κB activation and the downstream production of proinflammatory mediators, whereas ectopic TLR4 expression significantly abrogated the suppressed inflammatory response induced by miR-181c. Therefore, our study identified an important role for the miR-181c-TLR4 pathway in hypoxic microglial activation and neuroinflammation. This pathway could represent a potential therapeutic target for cerebral hypoxic diseases associated with microglial activation and the inflammatory response. Cerebral hypoxia/ischemia induces microglial activation and the release of inflammatory cytokines. We found that hypoxia down-regulated miR-181c in primary microglia. In addition, miR-181c inhibited TLR4 expression through binding to its 3'UTR, thus inhibiting NF-kB activation and the production of downstream proinflammatory mediators. Therefore, the miR-181c-TLR4 pathway may be a potential therapeutic target for the treatment of cerebral hypoxic diseases.


Asunto(s)
Glucosa/deficiencia , Mediadores de Inflamación/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Oxígeno/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Femenino , Redes Reguladoras de Genes/fisiología , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4/antagonistas & inhibidores
4.
J Vasc Access ; : 11297298231158670, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36895143

RESUMEN

BACKGROUND: The risk factors for skin injuries remain poorly understood in cancer patients with peripherally inserted central catheters (PICC). We herein aimed at exploring the effect of clinical factors on the risk of PICC-related skin injuries. METHODS: We included 1245 cancer patients with PICC from 16 hospitals in Suzhou, China. The study outcome was in-hospital skin injuries, including contact dermatitis, skin (epidermal) stripping, tension injury, allergic dermatitis, skin tear, maceration, folliculitis, and pressure injury. RESULTS: During hospitalization, 274 patients (22.0%) developed skin injuries after prolonged use of an indwelling catheter. Univariable logistic regression analysis identified several risk factors for PICC-related skin injuries; multivariable logistic regression analysis showed that the following factors independently and significantly (p < 0.05) associated with the risk of PICC-related skin injuries: body mass index (BMI, >25 kg/m2 versus <18.5 kg/m2: odds ratio (OR), 1.79; 95% confidence interval (CI), 1.03-3.11), skin condition (humid vs normal: OR, 2.96; 95% CI, 1.62-5.43), skin indentation (OR, 4.67; 95% CI, 3.31-6.58), allergic history (OR, 2.11; 95% CI, 1.21-3.66), history of dermatitis (OR, 3.05; 95% CI, 1.00-9.28), history of eczema (OR, 3.36; 95% CI, 1.20-9.43), catheter insertion site (under elbow vs. upper arm: OR, 3.32; 95% CI, 1.12-9.90), and PICC maintenance interval (4-5 days vs ⩽3 days: OR, 0.06; 95% CI, 0.01-0.50; 5-7 days vs ⩽3 days: OR, 0.07; 95% CI, 0.02-0.31; 7-9 days vs ⩽3 days: OR, 0.10; 95% CI, 0.02-0.57). CONCLUSIONS: BMI, skin condition, skin indentation, allergic history, history of dermatitis, history of eczema, catheter insertion site, and PICC maintenance interval were independent risk factors for PICC-related skin injuries in cancer patients. This knowledge will guide future studies with formulating optimal treatment strategies for improving the skin health of cancer patients with PICC.

5.
Glia ; 60(12): 1888-95, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22907769

RESUMEN

A growing body of evidence suggests that microRNA (miRNA) dysregulation contributes to many types of human disease, including central nervous system disorders. In this study, we identified an inverse correlation between the expression of miR-21 and Fas ligand (FasL) during hypoxia-induced microglial activation. Specifically, hypoxia caused the upregulation of FasL expression but the downregulation of miR-21 expression in microglia. Furthermore, we demonstrated that miR-21 suppresses FasL production by directly binding to its 3'-untranslated region. The overproduction of FasL following hypoxic microglial activation induced neuronal apoptosis, whereas the ectopic expression of miR-21 partially protected neurons from cell death caused by hypoxia-activated microglia. Finally, we confirmed that the function of miR-21 in microglia-mediated neuronal injury is dependent on FasL. Our study demonstrates an important role for miRNAs in microglia-mediated neuronal apoptosis, and suggests potential novel therapeutic interventions for cerebral hypoxic diseases associated with microglial activation.


Asunto(s)
Proteína Ligando Fas/antagonistas & inhibidores , MicroARNs/fisiología , Microglía/fisiología , Neuronas/patología , Animales , Animales Recién Nacidos , Muerte Celular/fisiología , Hipoxia de la Célula/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Proteína Ligando Fas/biosíntesis , Proteína Ligando Fas/genética , Células HEK293 , Humanos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/fisiología
6.
J Neuroinflammation ; 9: 211, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22950459

RESUMEN

BACKGROUND: Post-ischemic microglial activation may contribute to neuronal damage through the release of large amounts of pro-inflammatory cytokines and neurotoxic factors. The involvement of microRNAs (miRNAs) in the pathogenesis of disorders related to the brain and central nervous system has been previously studied, but it remains unknown whether the production of pro-inflammatory cytokines is regulated by miRNAs. METHODS: BV-2 and primary rat microglial cells were activated by exposure to oxygen-glucose deprivation (OGD). Global cerebral ischemia was induced using the four-vessel occlusion (4-VO) model in rats. Induction of pro-inflammatory and neurotoxic factors, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and nitric oxide (NO), were assessed by ELISA, immunofluorescence, and the Griess assay, respectively. The miRNA expression profiles of OGD-activated BV-2 cells were subsequently compared with the profiles of resting cells in a miRNA microarray. BV-2 and primary rat microglial cells were transfected with miR-181c to evaluate its effects on TNF-α production after OGD. In addition, a luciferase reporter assay was conducted to confirm whether TNF-α is a direct target of miR-181c. RESULTS: OGD induced BV-2 microglial activation in vitro, as indicated by the overproduction of TNF-α, IL-1ß, and NO. Global cerebral ischemia/reperfusion injury induced microglial activation and the release of pro-inflammatory cytokines in the hippocampus. OGD also downregulated miR-181c expression and upregulated TNF-α expression. Overproduction of TNF-α after OGD-induced microglial activation provoked neuronal apoptosis, whereas the ectopic expression of miR-181c partially protected neurons from cell death caused by OGD-activated microglia. RNAinterference-mediated knockdown of TNF-α phenocopied the effect of miR-181c-mediated neuronal protection, whereas overexpression of TNF-α blocked the miR-181c-dependent suppression of apoptosis. Further studies showed that miR-181c could directly target the 3'-untranslated region of TNF-α mRNA, suppressing its mRNA and protein expression. CONCLUSIONS: Our data suggest a potential role for miR-181c in the regulation of TNF-α expression after ischemia/hypoxia and microglia-mediated neuronal injury.


Asunto(s)
Apoptosis/fisiología , Isquemia Encefálica/patología , MicroARNs/metabolismo , Microglía/fisiología , Neuronas/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Isquemia Encefálica/metabolismo , Antígeno CD11b , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucosa/deficiencia , Hipocampo/citología , Hipoxia , Neuronas/efectos de los fármacos , Óxido Nítrico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Transfección
7.
Neuropathology ; 31(2): 112-21, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20667013

RESUMEN

Vascular factors have been shown to be important in cognitive impairment and dementia in the elderly. Recent evidence suggests that treatment at the stage of mild cognitive impairment (MCI) can prevent progression to dementia. In this study we established a rat model that simulates the pathophysiological condition of vascular MCI, characterized by gait disturbance in the absence of motor deficits and mild working memory dysfunction and not being demented. Initiation of vascular MCI pathology was not associated with loss of neurons, but was correlated with microglial activation and white matter changes. This MCI rat model will be useful for analysis of effects of vascular factors on cognitive dysfunction and neurodegenerative processes and development of drugs for treatment of this disorder.


Asunto(s)
Circulación Cerebrovascular/fisiología , Trastornos Cerebrovasculares/fisiopatología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Animales , Trastornos Cerebrovasculares/complicaciones , Trastornos Cerebrovasculares/patología , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Cojera Animal/etiología , Cojera Animal/fisiopatología , Flujometría por Láser-Doppler , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Ratas , Ratas Wistar
8.
Front Cell Neurosci ; 12: 154, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29937716

RESUMEN

Background: Microglia play an important role in the central nervous system as immune cells and are often activated by post-ischemic injury. MicroRNAs are small endogenous RNAs affecting many complex cellular biological functions that are involved in neurodegenerative and cerebrovascular diseases. Previous studies have shown that microRNA-26b (miR-26b) is downregulated in BV-2 cells exposed to oxygen-glucose deprivation (OGD). Objective: This study aimed to investigate how miR-26b regulates microglial activation and its neurotoxicity as well as the effect of miR-26b on vascular cognitive impairment (VCI). Methods: Here, we used PCR to detect the mRNA expression of miR-26b and cytokines, western blot for the protein expression of cytokines, and the live/dead assay for neuronal apoptosis. In addition, we employed a luciferase assay to identify the possible target genes of miR-26b. Furthermore, we studied the effects of cerebral ischemia by bilateral common carotid artery occlusion (BCCAO) in rats. We used staining to identify neurons and microglia, and we tested cognitive function by the T-maze test. Results: Our results showed that OGD activated microglia and increased the expression of interleukin (IL)-6 and other cytokines. Similarly, BCCAO activated microglia and increased the expression of IL-6 in the hippocampal CA1 area. We further found that miR-26b decreased the number of activated microglia and targeted IL-6. Moreover, miR-26b expression attenuated microglial activation, inflammation, neurotoxicity and VCI. Conclusion: Our results suggested that miR-26b is involved in microglial activation and neurotoxicity in hypoxia/ischemia via IL-6. Therefore, increasing miR-26b expression may improve cognitive function.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA