Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Virol J ; 21(1): 89, 2024 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641810

RESUMEN

Coxsackievirus-A10 (CV-A10), responsible for the hand, foot and mouth disease (HFMD) pandemic, could cause serious central nervous system (CNS) complications. The underlying molecular basis of CV-A10 and host interactions inducing neuropathogenesis is still unclear. The Hippo signaling pathway, historically known for a dominator of organ development and homeostasis, has recently been implicated as an immune regulator. However, its role in host defense against CV-A10 has not been investigated. Herein, it was found that CV-A10 proliferated in HMC3 cells and promoted the release of inflammatory cytokines. Moreover, pattern recognition receptors (PRRs)-mediated pathways, including TLR3-TRIF-TRAF3-TBK1-NF-κB axis, RIG-I/MDA5-MAVS-TRAF3-TBK1-NF-κB axis and TLR7-MyD88-IRAK1/IRAK4-TRAF6-TAK1-NF-κB axis, were examined to be elevated under CV-A10 infection. Meanwhile, it was further uncovered that Hippo signaling pathway was inhibited in HMC3 cells with CV-A10 infection. Previous studies have been reported that there exist complex relations between innate immune and Hippo signaling pathway. Then, plasmids of knockdown and overexpression of MST1/2 were transfected into HMC3 cells. Our results showed that MST1/2 suppressed the levels of inflammatory cytokines via interacting with TBK1 and IRAK1, and also enhanced virus production via restricting IRF3 and IFN-ß expressions. Overall, these data obviously pointed out that CV-A10 accelerated the formation of neuroinflammation by the effect of the Hippo pathway on the PRRs-mediated pathway, which delineates a negative immunoregulatory role for MST1/2 in CV-A10 infection and the potential for this pathway to be pharmacologically targeted to treat CV-A10.


Asunto(s)
Bencenoacetamidas , Infecciones por Coxsackievirus , FN-kappa B , Piperidonas , Humanos , FN-kappa B/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Enfermedades Neuroinflamatorias , Inmunidad Innata , Citocinas/metabolismo
2.
Virol J ; 20(1): 178, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37559147

RESUMEN

Coxsackievirus A16 (CV-A16) is still an important pathogen that causes hand, foot and mouth disease (HFMD) in young children and infants worldwide. Previous studies indicated that CV-A16 infection is usually mild or self-limiting, but it was also found that CV-A16 infection can trigger severe neurological complications and even death. However, there are currently no vaccines or antiviral compounds available to either prevent or treat CV-A16 infection. Therefore, investigation of the virus‒host interaction and identification of host proteins that play a crucial regulatory role in the pathogenesis of CV-A16 infection may provide a novel strategy to develop antiviral drugs. Here, to increase our understanding of the interaction of CV-A16 with the host cell, we analyzed changes in the proteome of 16HBE cells in response to CV-A16 using tandem mass tag (TMT) in combination with LC‒MS/MS. There were 6615 proteins quantified, and 172 proteins showed a significant alteration during CV-A16 infection. These differentially regulated proteins were involved in fundamental biological processes and signaling pathways, including metabolic processes, cytokine‒cytokine receptor interactions, B-cell receptor signaling pathways, and neuroactive ligand‒receptor interactions. Further bioinformatics analysis revealed the characteristics of the protein domains and subcellular localization of these differentially expressed proteins. Then, to validate the proteomics data, 3 randomly selected proteins exhibited consistent changes in protein expression with the TMT results using Western blotting and immunofluorescence methods. Finally, among these differentially regulated proteins, we primarily focused on HMGB1 based on its potential effects on viral replication and virus infection-induced inflammatory responses. It was demonstrated that overexpression of HMGB1 could decrease viral replication and upregulate the release of inflammatory cytokines, but deletion of HMGB1 increased viral replication and downregulated the release of inflammatory cytokines. In conclusion, the results from this study have helped further elucidate the potential molecular pathogenesis of CV-A16 based on numerous protein changes and the functions of HMGB1 Found to be involved in the processes of viral replication and inflammatory response, which may facilitate the development of new antiviral therapies as well as innovative diagnostic methods.


Asunto(s)
Enterovirus , Proteína HMGB1 , Replicación Viral , Humanos , Cromatografía Liquida , Citocinas/metabolismo , Enterovirus/fisiología , Enfermedad de Boca, Mano y Pie , Proteína HMGB1/metabolismo , Proteómica , Espectrometría de Masas en Tándem , Línea Celular
3.
Onco Targets Ther ; 14: 1807-1819, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33727831

RESUMEN

BACKGROUND: Circular RNA (circRNA) is a key regulator of cancer, and it has been proved to be involved in the regulation of cancer progression including non-small cell lung cancer (NSCLC). Circ-PITX1 was found to be a significantly upregulated circRNA in NSCLC, and its role and potential mechanism in NSCLC progression deserve further investigation. METHODS: The expression levels of circ-PITX1, microRNA (miR)-1248 and cyclin D2 (CCND2) were examined by quantitative real-time PCR (qRT-PCR). Cell proliferation, apoptosis, cell cycle process, migration and invasion were determined using cell counting kit 8 (CCK8) assay, colony formation assay, flow cytometry, wound healing assay and transwell assay. Xenograft models were built to explore the role of circ-PITX1 in NSCLC tumor growth in vivo. The glycolysis and glutamine metabolism of cells were assessed by detecting the consumptions of glucose and glutamine, cell extracellular acidification rate (ECAR), and the productions of lactate, α-ketoglutaric acid (α-KG) and ATP. The protein levels of hexokinase 2 (HK-2), glutaminase 1 (GLS1) and CCND2 were tested by Western blot (WB) analysis. Dual-luciferase reporter assay and RIP assay were employed to verify the interaction between miR-1248 and circ-PITX1 or CCND2. RESULTS: Circ-PITX1 was upregulated in NSCLC and its silencing could inhibit the proliferation, migration, invasion, cell cycle process, glycolysis, glutamine metabolism, and promote the apoptosis of NSCLC cells in vitro, as well as reduced tumor growth in vivo. In the terms of mechanism, we found that circ-PITX1 could act as a sponge of miR-1248, and miR-1248 could target CCND2. In addition, miR-1248 inhibitor reversed the inhibitory effect of circ-PITX1 knockdown on NSCLC progression. Similarly, CCND2 overexpression also reversed the suppressive effect of miR-1248 on NSCLC progression. Moreover, circ-PITX1 positively regulated CCND2 expression by sponging miR-1248. CONCLUSION: Circ-PITX1 served as a sponge of miR-1248 to promote NSCLC progression by upregulating CCND2.

4.
Sci Rep ; 6: 22884, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26964899

RESUMEN

Acetylcholine receptors (AChRs) serve as connections between motor neurons and skeletal muscle and are essential for recovery from spinal cord transection (SCT). Recently, microRNAs have emerged as important potential biotherapeutics for several diseases; however, whether miRNAs operate in the modulation of AChRs remains unknown. We found increased AChRs numbers and function scores in rats with SCT; these increases were reduced following the injection of a eukaryotic translation initiation factor 5A1 (eIF5A1) shRNA lentivirus into the hindlimb muscle. Then, high-throughput screening for microRNAs targeting eIF5A1 was performed, and miR-434-3p was found to be robustly depleted in SCT rat skeletal muscle. Furthermore, a highly conserved miR-434-3p binding site was identified within the mRNA encoding eIF5A1 through bioinformatics analysis and dual-luciferase assay. Overexpression or knockdown of miR-434-3p in vivo demonstrated it was a negative post-transcriptional regulator of eIF5A1 expression and influenced AChRs expression. The microarray-enriched Gene Ontology (GO) terms regulated by miR-434-3p were muscle development terms. Using a lentivirus, one functional gene (map2k6) was confirmed to have a similar function to that of miR-434-3p in GO terms. Finally, HRM and MeDIP-PCR analyses revealed that DNA demethylation also up-regulated eIF5A1 after SCT. Consequently, miR-434-3p/eIF5A1 in muscle is a promising potential biotherapy for SCI repair.


Asunto(s)
Metilación de ADN , Regulación de la Expresión Génica , MicroARNs/genética , Músculo Esquelético/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Receptores Nicotínicos/genética , Traumatismos de la Médula Espinal/genética , Regiones no Traducidas 3' , Animales , Sitios de Unión , Biología Computacional , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ontología de Genes , Anotación de Secuencia Molecular , Actividad Motora , Interferencia de ARN , Ratas , Traumatismos de la Médula Espinal/rehabilitación , Transcriptoma , Factor 5A Eucariótico de Iniciación de Traducción
5.
Mol Neurobiol ; 52(1): 522-32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25204493

RESUMEN

Spinal cord injury (SCI) results in a series of severe dysfunction of sensory and motor functions, while the molecular mechanisms that cause these dysfunctions remain elusive. Using proteomics technology, Western blot (WB), and immunohistochemistry (IHC), we found endoplasmic reticulum protein 29 (ERp29) was substantially downregulated in the motor cortex 3 days postoperation (dpo) after spinal cord transection (SCT, T10) followed by a gradual recovery 28 dpo. IHC showed that ERp29 is expressed in cortical neurons. In order to investigate the role of ERp29 in axotomized cortical neurons, we developed an in vitro axotomy injury model. ERp29 overexpression in cortical neurons after axotomy protected them from apoptosis; prevented the reduction of the number of neurons, and prevented reduction of neurite length. Moreover, we found that ERp29 overexpression increased neuronal regeneration assessed by neurite number and length. Furthermore, overexpression of ERp29 in cortical neurons after axotomy increased expression of Erk-1 and PI3K while decreasing the expression of caspase-3 expression. The present data therefore provides evidence to address the role of ERp29 in axotomized cortical neurons and identifies new therapeutic targets for the treatment of SCI.


Asunto(s)
Apoptosis , Axotomía , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Choque Térmico/metabolismo , Regeneración Nerviosa , Neuronas/metabolismo , Neuroprotección , Animales , Caspasa 3/metabolismo , Supervivencia Celular , Corteza Cerebral/metabolismo , Electroforesis en Gel Bidimensional , Femenino , Sistema de Señalización de MAP Quinasas , Neuritas/metabolismo , Proteómica , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Factores de Tiempo
6.
Mol Neurobiol ; 51(2): 437-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24874717

RESUMEN

Spinal cord injury (SCI) often results in motor disability concomitant with limit neuroplasticity; the underlying mechanism, however, is still unclear. This study established spinal cord transection rats model (T10), then performed cDNA microarray analysis and found that vimentin located in astrocytes was increased significantly in scar tissues after transection. To understand the role of vimentin and it's mechanism of regulation, RNA interference and luciferase assay were used. Vimentin knockdown in the scar tissues showed a significant improvement on locomotor function in hindlimbs, while vimentin overexpression exhibited an opposite effect. In vitro, vimentin downregulation or overexpression can effectively inhibit or increase astrogliosis, respectively. Moreover, by using biological informatics technology, we predicted that vimentin may be as the target of micRNA138 (miR-138), and confirmed that miR-138 could regulate vimentin by luciferase activity assay. The present results not only validated the exact role of vimentin in transected spinal cord, but also exhibited a novel regulation mechanism, in which miR-138 may regulate vimentin to promote neuroplasticity. It, therefore, provides a novel target for gene drug discovery based on miRNA-138 or vimentin for the treatment of SCI in the future clinic trial.


Asunto(s)
MicroARNs/biosíntesis , Plasticidad Neuronal/fisiología , Traumatismos de la Médula Espinal/metabolismo , Vimentina/fisiología , Animales , Animales Recién Nacidos , Secuencia de Bases , Células Cultivadas , Femenino , Masculino , MicroARNs/genética , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología
7.
Mol Neurobiol ; 50(2): 290-304, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24519330

RESUMEN

Spinal cord injury causes sensory loss below the level of lesion. Synaptosomal-associated protein 25 (SNAP25) is a t-SNARE protein essential for exocytosis and neurotransmitter release, but its role in sensory functional recovery has not been determined. The aim of the present study is therefore to investigate whether SNAP25 can promote sensory recovery. By 2D proteomics, we found a downregulation of SNAP25 and then constructed two lentiviral vectors, Lv-exSNAP25 and Lv-shSNAP25, which allows efficient and stable RNAi-mediated silencing of endogenous SNAP25. Overexpression of SNAP25 enhanced neurite outgrowth in vitro and behavior response to thermal and mechanical stimuli in vivo, while the silencing of SNAP25 had the opposite effect. These results suggest that SNAP25 plays a crucial role in sensory functional recovery following spinal cord injury (SCI). Our study therefore provides a novel target for the management of SCI for sensory dysfunction.


Asunto(s)
Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Exocitosis/fisiología , Masculino , Ratas Sprague-Dawley
8.
BMC Biochem ; 14: 21, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23914775

RESUMEN

BACKGROUND: Transforming growth factor-betas (TGF-ßs), including beta2 (TGF-ß2), constitute a superfamily of multifunctional cytokines with important implications in morphogenesis, cell differentiation and tissue remodeling. TGF-ß2 is thought to play important roles in multiple developmental processes and neuron survival. However, before we carried out these investigations, a TGF-ß2 gene down-regulated transgenic animal model was needed. In the present study, expressional silencing TGF-ß2 was achieved by select predesigning interference short hairpin RNAs (shRNAs) targeting mouse TGF-ß2 genes. RESULTS: Four homozygous transgenic offspring were generated by genetic manipulation and the protein expressions of TGF-ß2 were detected in different tissues of these mice. The transgenic mice were designated as Founder 66, Founder 16, Founder 53 and Founder 41. The rates of TGF-ß2 down-expression in different transgenic mice were evaluated. The present study showed that different TGF-ß2 expressions were detected in multiple tissues and protein levels of TGF-ß2 decreased at different rates relative to that of wild type mice. The expressions of TGF-ß2 proteins in transgenic mice (Founder 66) reduced most by 52%. CONCLUSIONS: The present study generated transgenic mice with TGF-ß2 down-regulated, which established mice model for systemic exploring the possible roles of TGF-ß2 in vivo in different pathology conditions.


Asunto(s)
Factor de Crecimiento Transformador beta2/metabolismo , Animales , Regulación de la Expresión Génica , Genotipo , Homocigoto , Ratones , Ratones Transgénicos , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Crecimiento Transformador beta2/antagonistas & inhibidores , Factor de Crecimiento Transformador beta2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...