Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Virol ; 95(2): e28574, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36772841

RESUMEN

Human cytomegalovirus (HCMV) preferentially targets neural progenitor cells (NPCs) in congenitally infected fetal brains, inducing neurodevelopmental disorders. While HCMV expresses several microRNAs (miRNAs) during infection, their roles in NPC infection are unclear. Here, we characterized expression of cellular and viral miRNAs in HCMV-infected NPCs during early infection by microarray and identified seven differentially expressed cellular miRNAs and six significantly upregulated HCMV miRNAs. Deep learning approaches were used to identify potential targets of significantly upregulated HCMV miRNAs against differentially expressed cellular messenger RNA (mRNAs), and the associations with miRNA-mRNA expression changes were observed. Gene ontology enrichment analysis indicated cellular gene targets were significantly enriched in pathways involved in neurodevelopment and cell-cycle processes. Viral modulation of selected miRNAs and cellular gene targets involved in neurodevelopmental processes were further validated by real-time quantitative reverse transcription polymerase chain reaction. Finally, a predicted 3' untranslated region target site of hcmv-miR-US25-1 in Jag1, a factor important for neurogenesis, was confirmed by mutagenesis. Reduction of Jag1 RNA and protein levels in NPCs was observed in response to transient expression of hcmv-miR-US25-1. A hcmv-miR-US25-1 mutant virus (ΔmiR-US25) displayed limited ability to downregulate Jag1 mRNA levels and protein levels during the early infection stage compared with the wild type virus. Our collective experimental and computational investigation of miRNAs and cellular mRNAs expression in HCMV-infected NPCs yields new insights into the roles of viral miRNAs in regulating NPC fate and their contributions to HCMV neuropathogenesis.


Asunto(s)
Infecciones por Citomegalovirus , MicroARNs , Humanos , MicroARNs/genética , Citomegalovirus/genética , Células Madre/metabolismo
2.
Nature ; 579(7797): 118-122, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103178

RESUMEN

It has long been assumed that lifespan and healthspan correlate strongly, yet the two can be clearly dissociated1-6. Although there has been a global increase in human life expectancy, increasing longevity is rarely accompanied by an extended healthspan4,7. Thus, understanding the origin of healthy behaviours in old people remains an important and challenging task. Here we report a conserved epigenetic mechanism underlying healthy ageing. Through genome-wide RNA-interference-based screening of genes that regulate behavioural deterioration in ageing Caenorhabditis elegans, we identify 59 genes as potential modulators of the rate of age-related behavioural deterioration. Among these modulators, we found that a neuronal epigenetic reader, BAZ-2, and a neuronal histone 3 lysine 9 methyltransferase, SET-6, accelerate behavioural deterioration in C. elegans by reducing mitochondrial function, repressing the expression of nuclear-encoded mitochondrial proteins. This mechanism is conserved in cultured mouse neurons and human cells. Examination of human databases8,9 shows that expression of the human orthologues of these C. elegans regulators, BAZ2B and EHMT1, in the frontal cortex increases with age and correlates positively with the progression of Alzheimer's disease. Furthermore, ablation of Baz2b, the mouse orthologue of BAZ-2, attenuates age-dependent body-weight gain and prevents cognitive decline in ageing mice. Thus our genome-wide RNA-interference screen in C. elegans has unravelled conserved epigenetic negative regulators of ageing, suggesting possible ways to achieve healthy ageing.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Epigénesis Genética , Envejecimiento Saludable/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Factores Generales de Transcripción/metabolismo , Envejecimiento/genética , Animales , Proteínas de Caenorhabditis elegans/genética , Cognición , Disfunción Cognitiva , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/metabolismo , Humanos , Longevidad/genética , Lisina/metabolismo , Masculino , Memoria , Metilación , Ratones , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteínas/genética , Interferencia de ARN , Aprendizaje Espacial , Factores Generales de Transcripción/deficiencia , Factores Generales de Transcripción/genética
3.
Medicine (Baltimore) ; 98(25): e16109, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31232956

RESUMEN

RATIONALE: It is rare to find 22q11.2 deletion syndrome with pseudohypoparathyroidism in children. Furthermore, the phenotypic spectrum of this disorder varies widely. PATIENT CONCERNS: A patient was diagnosed with pseudohypoparathyroidism at age 14 years because of convulsions, hypocalcemia, hyperphosphatemia, normal parathyroid hormone levels, and basal ganglia calcifications. Thereafter, the child presented with symptoms of nephrotic syndrome; subsequently, he was diagnosed with nephrotic syndrome at the local hospital. DIAGNOSIS: At our hospital, multiplex ligation-dependent probe amplification confirmed that the patient had 22q11.2 deletion syndrome. INTERVENTIONS: The patient continued to be treated with calcium supplements. OUTCOMES: Seizure activity and proteinuria ceased. LESSONS: Signs of this syndrome include delayed speech development due to velofacial dysfunction, recurrent croup attacks during early childhood due to latent hypocalcemia, and mild dysmorphic features. The findings of this patient indicated that 22q11.2 deletion syndrome may include a wide spectrum of clinical findings and that this diagnosis needs to be considered for all patients presenting with hypocalcemia, regardless of age.


Asunto(s)
Síndrome de DiGeorge/diagnóstico , Seudohipoparatiroidismo/diagnóstico , Adolescente , Síndrome de DiGeorge/genética , Humanos , Hiperfosfatemia/etiología , Hipocalcemia/etiología , Masculino , Reacción en Cadena de la Polimerasa Multiplex/métodos , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/genética , Convulsiones/etiología
4.
Cancer Cell Int ; 18: 149, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30275772

RESUMEN

BACKGROUND: Marsdenia tenacissima is an herb medicine which has been utilized to treat malignant diseases for decades. The M. tenacissima extract (MTE) shows significant anti-proliferation activity against non-small cell lung cancer (NSCLC) cells, but the underlying mechanisms remain unclear. In this study, we explored the potential anti-proliferation mechanisms of MTE in NSCLC cells in relation to apoptosis as well as autophagy, which are two critical forms to control cancer cell survival and death. METHODS: The proliferation of H1975 and A549 cells was evaluated by MTT assay. Cell apoptosis was assessed by Annexin V and PI staining, Caspase 3 expression and activity. Autophagy flux proteins were detected by Western blot with or without autophagy inducer and inhibitor. Endogenous LC3-II puncta and LysoTracker staining were monitored by confocal microscopy. The formation of autophagic vacuoles was measured by acridine orange staining. ERK is a crucial molecule to interplay with cell autophagy and apoptosis. The role of ERK on cell apoptosis and autophagy influenced by MTE was determined in the presence of MEK/ERK inhibitor U0126. RESULTS: The significant growth inhibition and apoptosis induction were observed in MTE treated NSCLC cells. MTE induced cell apoptosis coexisted with elevated Caspase 3 activity. MTE also impaired autophagic flux by upregulated LC3-II and p62 expression. Autophagy inducer EBSS could not abolish the impaired autophagic flux by MTE, while it was augmented in the presence of autophagy inhibitor Baf A1. The autophagosome-lysosome fusion was blocked by MTE via affecting lysosome function as evidenced by decreased expression of LAMP1 and Cathepsin B. The molecule ERK became hyperactivated after MTE treatment, but the MEK/ERK inhibitor U0126 abrogated autophagy inhibition and apoptosis induction caused by MTE, suggested that ERK signaling pathways partially contributed to cell death caused by MTE. CONCLUSION: Our results demonstrate that MTE caused apoptosis induction as well as autophagy inhibition in NSCLC cells. The activated ERK is partially associated with NSCLC apoptotic and autophagic cell death in response to MTE treatment. The present findings reveal new mechanisms for the anti-tumor activity of MTE against NSCLC.

5.
Nat Commun ; 9(1): 3941, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30258187

RESUMEN

Ion channels are important therapeutic targets, but the discovery of ion channel drugs remains challenging due to a lack of assays that allow high-throughput screening in the physiological context. Here we report C. elegans phenotype-based methods for screening ion channel drugs. Expression of modified human ether-a-go-go-related gene (hERG) potassium channels in C. elegans results in egg-laying and locomotive defects, which offer indicators for screening small-molecule channel modulators. Screening in worms expressing hERGA561V, which carries a trafficking-defective mutation A561V known to associate with long-QT syndrome, identifies two functional correctors Prostratin and ingenol-3,20-dibenzoate. These compounds activate PKCε signaling and consequently phosphorylate S606 at the pore region of the channel to promote hERGA561V trafficking to the plasma membrane. Importantly, the compounds correct electrophysiological abnormalities in hiPSC-derived cardiomyocytes bearing a heterozygous CRISPR/Cas9-edited hERGA561V. Thus, we have developed an in vivo high-throughput method for screening compounds that have therapeutic potential in treating channelopathies.


Asunto(s)
Canalopatías/genética , Canales de Potasio Éter-A-Go-Go/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Canalopatías/tratamiento farmacológico , Canalopatías/metabolismo , Modelos Animales de Enfermedad , Diterpenos/farmacología , Diterpenos/uso terapéutico , Evaluación Preclínica de Medicamentos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ésteres del Forbol/farmacología , Ésteres del Forbol/uso terapéutico , Proteína Quinasa C/metabolismo , Triterpenos/farmacología , Triterpenos/uso terapéutico
6.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950413

RESUMEN

The mechanisms underlying neurodevelopmental damage caused by virus infections remain poorly defined. Congenital human cytomegalovirus (HCMV) infection is the leading cause of fetal brain development disorders. Previous work has linked HCMV infection to perturbations of neural cell fate, including premature differentiation of neural progenitor cells (NPCs). Here, we show that HCMV infection of NPCs results in loss of the SOX2 protein, a key pluripotency-associated transcription factor. SOX2 depletion maps to the HCMV major immediate early (IE) transcription unit and is individually mediated by the IE1 and IE2 proteins. IE1 causes SOX2 downregulation by promoting the nuclear accumulation and inhibiting the phosphorylation of STAT3, a transcriptional activator of SOX2 expression. Deranged signaling resulting in depletion of a critical stem cell protein is an unanticipated mechanism by which the viral major IE proteins may contribute to brain development disorders caused by congenital HCMV infection.IMPORTANCE Human cytomegalovirus (HCMV) infections are a leading cause of brain damage, hearing loss, and other neurological disabilities in children. We report that the HCMV proteins known as IE1 and IE2 target expression of human SOX2, a central pluripotency-associated transcription factor that governs neural progenitor cell (NPC) fate and is required for normal brain development. Both during HCMV infection and when expressed alone, IE1 causes the loss of SOX2 from NPCs. IE1 mediates SOX2 depletion by targeting STAT3, a critical upstream regulator of SOX2 expression. Our findings reveal an unanticipated mechanism by which a common virus may cause damage to the developing nervous system and suggest novel targets for medical intervention.


Asunto(s)
Citomegalovirus/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/virología , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/metabolismo , Células Cultivadas , Humanos
7.
J Virol ; 92(9)2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29437978

RESUMEN

WD repeat-containing protein 5 (WDR5) is essential for assembling the VISA-associated complex to induce a type I interferon antiviral response to Sendai virus infection. However, the roles of WDR5 in DNA virus infections are not well described. Here, we report that human cytomegalovirus exploits WDR5 to facilitate capsid nuclear egress. Overexpression of WDR5 in fibroblasts slightly enhanced the infectious virus yield. However, WDR5 knockdown dramatically reduced infectious virus titers with only a small decrease in viral genome replication or gene expression. Further investigation of late steps of viral replication found that WDR5 knockdown significantly impaired formation of the viral nuclear egress complex and induced substantially fewer infoldings of the inner nuclear membrane. In addition, fewer capsids were associated with these infoldings, and there were fewer capsids in the cytoplasm. Restoration of WDR5 partially reversed these effects. These results suggest that WDR5 knockdown impairs the nuclear egress of capsids, which in turn decreases virus titers. These findings reveal an important role for a host factor whose function(s) is usurped by a viral pathogen to promote efficient replication. Thus, WDR5 represents an interesting regulatory mechanism and a potential antiviral target.IMPORTANCE Human cytomegalovirus (HCMV) has a large (∼235-kb) genome with over 170 open reading frames and exploits numerous cellular factors to facilitate its replication. HCMV infection increases protein levels of WD repeat-containing protein 5 (WDR5) during infection, overexpression of WDR5 enhances viral replication, and knockdown of WDR5 dramatically attenuates viral replication. Our results indicate that WDR5 promotes the nuclear egress of viral capsids, the depletion of WDR5 resulting in a significant decrease in production of infectious virions. This is the first report that WDR5 favors HCMV, a DNA virus, replication and highlights a novel target for antiviral therapy.


Asunto(s)
Cápside/metabolismo , Citomegalovirus/fisiología , Replicación del ADN/genética , ADN Viral/biosíntesis , N-Metiltransferasa de Histona-Lisina/metabolismo , Replicación Viral/fisiología , Línea Celular , Supervivencia Celular , ADN Viral/genética , Genoma Viral/genética , Células HEK293 , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Pulmón/virología , Transporte de Proteínas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Regulación hacia Arriba , Carga Viral/genética , Internalización del Virus
8.
Nature ; 551(7679): 198-203, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29120414

RESUMEN

The rate of behavioural decline in the ageing population is remarkably variable among individuals. Despite the considerable interest in studying natural variation in ageing rate to identify factors that control healthy ageing, no such factor has yet been found. Here we report a genetic basis for variation in ageing rates in Caenorhabditis elegans. We find that C. elegans isolates show diverse lifespan and age-related declines in virility, pharyngeal pumping, and locomotion. DNA polymorphisms in a novel peptide-coding gene, named regulatory-gene-for-behavioural-ageing-1 (rgba-1), and the neuropeptide receptor gene npr-28 influence the rate of age-related decline of worm mating behaviour; these two genes might have been subjected to recent selective sweeps. Glia-derived RGBA-1 activates NPR-28 signalling, which acts in serotonergic and dopaminergic neurons to accelerate behavioural deterioration. This signalling involves the SIR-2.1-dependent activation of the mitochondrial unfolded protein response, a pathway that modulates ageing. Thus, natural variation in neuropeptide-mediated glia-neuron signalling modulates the rate of ageing in C. elegans.


Asunto(s)
Envejecimiento/genética , Envejecimiento/fisiología , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Variación Genética , Neuroglía/metabolismo , Neuronas/metabolismo , Transducción de Señal/genética , Alelos , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Genética de Población , Locomoción/genética , Locomoción/fisiología , Longevidad/genética , Longevidad/fisiología , Masculino , Faringe/fisiología , Polimorfismo de Nucleótido Simple/genética , Receptores Acoplados a Proteínas G/metabolismo , Neuronas Serotoninérgicas/metabolismo , Conducta Sexual Animal/fisiología , Sirtuinas/metabolismo , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
9.
PLoS Pathog ; 13(7): e1006542, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28750047

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is the leading cause of neurological disabilities in children worldwide, but the mechanisms underlying these disorders are far from well-defined. HCMV infection has been shown to dysregulate the Notch signaling pathway in human neural progenitor cells (NPCs). As an important downstream effector of Notch signaling, the transcriptional regulator Hairy and Enhancer of Split 1 (Hes1) is essential for governing NPC fate and fetal brain development. In the present study, we report that HCMV infection downregulates Hes1 protein levels in infected NPCs. The HCMV 72-kDa immediate-early 1 protein (IE1) is involved in Hes1 degradation by assembling a ubiquitination complex and promoting Hes1 ubiquitination as a potential E3 ubiquitin ligase, followed by proteasomal degradation of Hes1. Sp100A, an important component of PML nuclear bodies, is identified to be another target of IE1-mediated ubiquitination. A C-terminal acidic region in IE1, spanning amino acids 451 to 475, is required for IE1/Hes1 physical interaction and IE1-mediated Hes1 ubiquitination, but is dispensable for IE1/Sp100A interaction and ubiquitination. Our study suggests a novel mechanism linking downregulation of Hes1 protein to neurodevelopmental disorders caused by HCMV infection. Our findings also complement the current knowledge of herpesviruses by identifying IE1 as the first potential HCMV-encoded E3 ubiquitin ligase.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Células-Madre Neurales/metabolismo , Factor de Transcripción HES-1/genética , Ubiquitina-Proteína Ligasas/metabolismo , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Células-Madre Neurales/enzimología , Células-Madre Neurales/virología , Unión Proteica , Proteolisis , Factor de Transcripción HES-1/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
10.
Virology ; 510: 205-215, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28750324

RESUMEN

T98G cells have been shown to support long-term human cytomegalovirus (HCMV) genome maintenance without infectious virus release. However, it remains unclear whether these viral genomes could be reactivated. To address this question, a recombinant HCMV (rHCMV) containing a GFP gene was used to infect T98G cells, and the infected cells absent of infectious virus production were designated T98G-LrV. Upon dibutyryl cAMP plus IBMX (cAMP/IBMX) treatment, a serial of phenomena were observed, including GFP signal increase, viral genome replication, lytic genes expression and infectious viruses release, indicating the reactivation of HCMV in T98G-LrV cells from a latent status. Mechanistically, HCMV reactivation in the T98G-LrV cells induced by cAMP/IBMX was associated with the PKA-CREB signaling pathway. These results demonstrate that HCMV was latent in T98G-LrV cells and could be reactivated. The T98G-LrV cells represent an effective model for investigating the mechanisms of HCMV reactivation from latency in the context of neural cells.


Asunto(s)
Citomegalovirus/fisiología , Activación Viral , Latencia del Virus , 1-Metil-3-Isobutilxantina/metabolismo , Bucladesina/metabolismo , Línea Celular Tumoral , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Coloración y Etiquetado/métodos
11.
Virol Sin ; 32(3): 188-198, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28451898

RESUMEN

Human cytomegalovirus (HCMV) infection is a leading cause of birth defects, primarily affecting the central nervous system and causing its maldevelopment. As the essential downstream effector of Notch signaling pathway, Hes1, and its dynamic expression, plays an essential role on maintaining neural progenitor /stem cells (NPCs) cell fate and fetal brain development. In the present study, we reported the first observation of Hes1 oscillatory expression in human NPCs, with an approximately 1.5 hour periodicity and a Hes1 protein half-life of about 17 (17.6 ± 0.2) minutes. HCMV infection disrupts the Hes1 rhythm and down-regulates its expression. Furthermore, we discovered that depleting Hes1 protein disturbed NPCs cell fate by suppressing NPCs proliferation and neurosphere formation, and driving NPCs abnormal differentiation. These results suggested a novel mechanism linking disruption of Hes1 rhythm and down-regulation of Hes1 expression to neurodevelopmental disorders caused by congenital HCMV infection.


Asunto(s)
Citomegalovirus/fisiología , Interacciones Huésped-Patógeno , Células-Madre Neurales/fisiología , Células-Madre Neurales/virología , Factor de Transcripción HES-1/biosíntesis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Humanos
12.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28356523

RESUMEN

Although a varicella-zoster virus (VZV) vaccine has been used for many years, the neuropathy caused by VZV infection is still a major health concern. Open reading frame 7 (ORF7) of VZV has been recognized as a neurotropic gene in vivo, but its neurovirulent role remains unclear. In the present study, we investigated the effect of ORF7 deletion on VZV replication cycle at virus entry, genome replication, gene expression, capsid assembly and cytoplasmic envelopment, and transcellular transmission in differentiated neural progenitor cells (dNPCs) and neuroblastoma SH-SY5Y (dSY5Y) cells. Our results demonstrate that the ORF7 protein is a component of the tegument layer of VZV virions. Deleting ORF7 did not affect viral entry, viral genome replication, or the expression of typical viral genes but clearly impacted cytoplasmic envelopment of VZV capsids, resulting in a dramatic increase of envelope-defective particles and a decrease in intact virions. The defect was more severe in differentiated neuronal cells of dNPCs and dSY5Y. ORF7 deletion also impaired transmission of ORF7-deficient virus among the neuronal cells. These results indicate that ORF7 is required for cytoplasmic envelopment of VZV capsids, virus transmission among neuronal cells, and probably the neuropathy induced by VZV infection.IMPORTANCE The neurological damage caused by varicella-zoster virus (VZV) reactivation is commonly manifested as clinical problems. Thus, identifying viral neurovirulent genes and characterizing their functions are important for relieving VZV related neurological complications. ORF7 has been previously identified as a potential neurotropic gene, but its involvement in VZV replication is unclear. In this study, we found that ORF7 is required for VZV cytoplasmic envelopment in differentiated neuronal cells, and the envelopment deficiency caused by ORF7 deletion results in poor dissemination of VZV among neuronal cells. These findings imply that ORF7 plays a role in neuropathy, highlighting a potential strategy to develop a neurovirulence-attenuated vaccine against chickenpox and herpes zoster and providing a new target for intervention of neuropathy induced by VZV.


Asunto(s)
Herpesvirus Humano 3/fisiología , Neuronas/fisiología , Neuronas/virología , Proteínas del Envoltorio Viral/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Cápside/metabolismo , Diferenciación Celular , Línea Celular , Citoplasma/virología , Eliminación de Gen , Genoma Viral , Herpes Zóster/virología , Herpesvirus Humano 3/genética , Humanos , Neuroblastoma , Proteínas del Envoltorio Viral/genética , Virión , Internalización del Virus , Replicación Viral
14.
Oncotarget ; 7(4): 4817-28, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26716895

RESUMEN

OBJECTIVE: miR-215 was reported to be downregulated and functioned as a tumor suppressor in several cancers. In contrast, miR-215 was preferentially upregulated in gastric cancer (GC) according to our data. Thus, we studied the potential biological function of miR-215 in GC. METHODS: miR-215 expression was measured in 77 paired GC tissues and adjacent non-tumor tissues. Biological functions of miR-215 were analyzed using cell viability, colony formation, migration, invasion, cell cycle, apoptosis and luciferase assays as well as via tumorigenicity and metastasis analysis. RESULTS: miR-215 was significantly upregulated in 7 GC cell lines and 77 GC tissues compared to adjacent non-tumor tissues (P < 0.05), and miR-215 expression was greater in advanced GC (stage III/IV; P < 0.05). Ectopic expression of miR-215 in GES-1 and HGC-27 cells (low miR-215 expression) promoted cell growth, migration, invasion, and metastasis, and these were reversed in NCI-N87 cells (high miR-215 expression) after miR-215 downregulation. Potential target genes of miR-215 were predicted and RUNX1, a transcription factor and a tumor suppressor, was confirmed to be potential target according to luciferase studies. RUNX1 was downregulated in GC tissues compared to adjacent non-tumor tissues (P < 0.05), and RUNX1 reversed partial function of miR-215 in vitro. CONCLUSIONS: miR-215 promotes malignant progression of GC by targeting RUNX1, and RUNX1 can partially reverse miR-215 effects.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Neoplasias Hepáticas/secundario , MicroARNs/genética , Neoplasias Gástricas/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Ciclo Celular , Movimiento Celular , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Virol ; 89(13): 6792-804, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25903338

RESUMEN

UNLABELLED: Human cytomegalovirus (HCMV) infection of the developing fetus frequently results in major neural developmental damage. In previous studies, HCMV was shown to downregulate neural progenitor/stem cell (NPC) markers and induce abnormal differentiation. As Notch signaling plays a vital role in the maintenance of stem cell status and is a switch that governs NPC differentiation, the effect of HCMV infection on the Notch signaling pathway in NPCs was investigated. HCMV downregulated mRNA levels of Notch1 and its ligand, Jag1, and reduced protein levels and altered the intracellular localization of Jag1 and the intracellular effector form of Notch1, NICD1. These effects required HCMV gene expression and appeared to be mediated through enhanced proteasomal degradation. Transient expression of the viral tegument proteins of pp71 and UL26 reduced NICD1 and Jag1 protein levels endogenously and exogenously. Given the critical role of Notch signaling in NPC growth and differentiation, these findings reveal important mechanisms by which HCMV disturbs neural cell development in vitro. Similar events in vivo may be associated with HCMV-mediated neuropathogenesis during congenital infection in the fetal brain. IMPORTANCE: Congenital human cytomegalovirus (HCMV) infection is the leading cause of birth defects that primarily manifest as neurological disabilities. Neural progenitor cells (NPCs), key players in fetal brain development, are the most susceptible cell type for HCMV infection in the fetal brain. Studies have shown that NPCs are fully permissive for HCMV infection, which causes neural cell loss and premature differentiation, thereby perturbing NPC fate. Elucidation of virus-host interactions that govern NPC proliferation and differentiation is critical to understanding neuropathogenesis. The Notch signaling pathway is critical for maintaining stem cell status and functions as a switch for differentiation of NPCs. Our investigation into the impact of HCMV infection on this pathway revealed that HCMV dysregulates Notch signaling by altering expression of the Notch ligand Jag1, Notch1, and its active effector in NPCs. These results suggest a mechanism for the neuropathogenesis induced by HCMV infection that includes altered NPC differentiation and proliferation.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Infecciones por Citomegalovirus/patología , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Células-Madre Neurales/fisiología , Receptor Notch1/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo , Infecciones por Citomegalovirus/virología , Regulación de la Expresión Génica , Humanos , Proteína Jagged-1 , Células-Madre Neurales/virología , Estabilidad Proteica , Proteolisis , Proteínas Serrate-Jagged
16.
J Virol ; 89(2): 1070-82, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378484

RESUMEN

UNLABELLED: Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, primarily manifesting as neurological disorders. HCMV infection alters expression of cellular microRNAs (miRs) and induces cell cycle arrest, which in turn modifies the cellular environment to favor virus replication. Previous observations found that HCMV infection reduces miR-21 expression in neural progenitor/stem cells (NPCs). Here, we show that infection of NPCs and U-251MG cells represses miR-21 while increasing the levels of Cdc25a, a cell cycle regulator and known target of miR-21. These opposing responses to infection prompted an investigation of the relationship between miR-21, Cdc25a, and viral replication. Overexpression of miR-21 in NPCs and U-251MG cells inhibited viral gene expression, genome replication, and production of infectious progeny, while shRNA-knockdown of miR-21 in U-251MG cells increased viral gene expression. In contrast, overexpression of Cdc25a in U-251MG cells increased viral gene expression and production of infectious progeny and overcame the inhibitory effects of miR-21 overexpression. Three viral gene products-IE1, pp71, and UL26-were shown to inhibit miR-21 expression at the transcriptional level. These results suggest that Cdc25a promotes HCMV replication and elevation of Cdc25a levels after HCMV infection are due in part to HCMV-mediated repression of miR-21. Thus, miR-21 is an intrinsic antiviral factor that is modulated by HCMV infection. This suggests a role for miR-21 downregulation in the neuropathogenesis of HCMV infection of the developing CNS. IMPORTANCE: Human cytomegalovirus (HCMV) is a ubiquitous pathogen and has very high prevalence among population, especially in China, and congenital HCMV infection is a major cause for birth defects. Elucidating virus-host interactions that govern HCMV replication in neuronal cells is critical to understanding the neuropathogenesis of birth defects resulting from congenital infection. In this study, we confirm that HCMV infection downregulates miR-21 but upregulates Cdc25a. Further determined the negative effects of cellular miRNA miR-21 on HCMV replication in neural progenitor/stem cells and U-251MG glioblastoma/astrocytoma cells. More importantly, our results provide the first evidence that miR-21 negatively regulates HCMV replication by targeting Cdc25a, a vital cell cycle regulator. We further found that viral gene products of IE1, pp71, and UL26 play roles in inhibiting miR-21 expression, which in turn causes increases in Cdc25a and benefits HCMV replication. Thus, miR-21 appears to be an intrinsic antiviral factor that represents a potential target for therapeutic intervention.


Asunto(s)
Citomegalovirus/inmunología , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Células-Madre Neurales/inmunología , Células-Madre Neurales/virología , Replicación Viral , Fosfatasas cdc25/metabolismo , Células Cultivadas , Citomegalovirus/fisiología , Humanos
17.
J Neurosci ; 34(11): 3947-58, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24623772

RESUMEN

Aging is accompanied with behavioral and cognitive decline. Changes in the neurotransmitter level are associated with the age-related behavioral deterioration, but whether well-known longevity manipulations affect the function of neurotransmitter system in aging animals is largely unclear. Here we report that serotonin (5-HT) and dopamine (DA) level decrease with age in C. elegans. The reduction results in downregulation of the activity of neurons controlled by 5-HT/DA signaling, and deterioration of some important behaviors, including pharyngeal pumping, food-induced slowing responses, and male mating. Longevity manipulations differentially affect the age-related decline in neuronal level of 5-HT/DA. The reduction and resultant behavioral deterioration occur in long-lived worms with defective insulin signaling [daf-2(e1370), age-1(hx546)] or mitochondria function [isp-1(qm150), tpk-1(qm162)], but not in long-lived worms with dietary restriction eat-2(ad1116). A reduced expression level of dopa decarboxylase BAS-1, the shared enzyme for 5-HT/DA synthesis, is responsible for the decline in 5-HT/DA levels. RNAi assay revealed that the sustained 5-HT/DA level in neurons of aged eat-2(ad1116) worms requires PHA-4 and its effectors superoxide dismutases and catalases, suggesting the involvement of reactive oxygen species in the 5-HT/DA decline. Furthermore, we found that elevating 5-HT/DA ameliorates age-related deterioration of pharyngeal pumping, food-induced slowing responses, and male mating in both wild-type and daf-2(e1370) worms. Together, dietary restriction preserves healthy behaviors in aged worms at least partially by sustaining a high 5-HT/DA level, and elevating the 5-HT/DA level in wild-type and daf-2(e1370) worms improves their behaviors during aging.


Asunto(s)
Envejecimiento/metabolismo , Caenorhabditis elegans/fisiología , Dopamina/metabolismo , Neuronas/metabolismo , Serotonina/metabolismo , Envejecimiento/fisiología , Animales , Conducta Animal/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Dopamina/deficiencia , Organismos Hermafroditas , Longevidad/fisiología , Masculino , Modelos Animales , Fenómenos Fisiológicos del Sistema Nervioso/fisiología , Neuronas/fisiología , Estrés Oxidativo/fisiología , Serotonina/deficiencia , Conducta Sexual Animal/fisiología , Transducción de Señal/fisiología
18.
J Virol ; 87(20): 10968-79, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903847

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is the most frequent infectious cause of birth defects, primarily neurological disorders. Neural progenitor/stem cells (NPCs) are the major cell type in the subventricular zone and are susceptible to HCMV infection. In culture, the differentiation status of NPCs may change with passage, which in turn may alter susceptibility to virus infection. Previously, only early-passage (i.e., prior to passage 9) NPCs were studied and shown to be permissive to HCMV infection. In this study, NPC cultures derived at different gestational ages were evaluated after short (passages 3 to 6) and extended (passages 11 to 20) in vitro passages for biological and virological parameters (i.e., cell morphology, expression of NPC markers and HCMV receptors, viral entry efficiency, viral gene expression, virus-induced cytopathic effect, and release of infectious progeny). These parameters were not significantly influenced by the gestational age of the source tissues. However, extended-passage cultures showed evidence of initiation of differentiation, increased viral entry, and more efficient production of infectious progeny. These results confirm that NPCs are fully permissive for HCMV infection and that extended-passage NPCs initiate differentiation and are more permissive for HCMV infection. Later-passage NPCs being differentiated and more permissive for HCMV infection suggest that HCMV infection in fetal brain may cause more neural cell loss and give rise to severe neurological disabilities with advancing brain development.


Asunto(s)
Encéfalo/citología , Citomegalovirus/crecimiento & desarrollo , Células-Madre Neurales/fisiología , Células-Madre Neurales/virología , Diferenciación Celular , Humanos , Pase Seriado
19.
Beijing Da Xue Xue Bao Yi Xue Ban ; 42(4): 480-4, 2010 Aug 18.
Artículo en Chino | MEDLINE | ID: mdl-20721270

RESUMEN

OBJECTIVE: To discuss a simple method for flow cytometric analysis of cell DNA stained with DAPI and Hoechst33342. METHODS: HT 29 cells stained with DAPI, Hoechst33342 or PI were measured by BD FACSAria and the percentages of cells in G0/G1, S and G2/M phases with three staining methods, then the results were analyzed and compared. Before measurement we monitored the quality of DNA analysis of flow cytometer through UV beads QC experiment and analyzed the standard chicken erythrocyte nuclei (CEN) and calf thymocyte nuclei (CTN) stained with DAPI and Hoechst33342. RESULTS: CV value of UV peak was 2.4 after QC experiments. There were 4 peaks on CEN histograms and the ratios of peak channel mean of G2/G1, G3/G1, and G4/G1 were about 2, 3, and 4 respectively. Both CV values of the first peak were 2.4. There were 2 peaks on CTN histograms and the ratio of peak channel mean of G2/G1 was 1.97, and CV value of G0/G1 2.4. The complete cell cycle of HT29 cells stained with DAPI, Hoechst33342 or PI was showed entirely, CV values were 3.40, 3.02 and 4.42, respectively, and the percentages of cells in G0/G1 were 60.86%, 60.22% and 60.81%,respectively, in S, 28.85%, 29.70% and 29.82%,respectively, and in G2/M, 10.29%, 9.09% and 9.37%, respectively. The results by the three methods showed no difference. CONCLUSION: This method for measurement of cellular DNA content is a simple and efficient approach to determining cell cycle and can be the first choice when using flow cytometer with 355 nm UV.


Asunto(s)
Bencimidazoles , ADN/análisis , Citometría de Flujo/métodos , Indoles , Ciclo Celular , Colorantes Fluorescentes , Células HT29 , Humanos
20.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 22(4): 214-6, 2010 Apr.
Artículo en Chino | MEDLINE | ID: mdl-20398465

RESUMEN

OBJECTIVE: To study the influence of diazoxide on mitochondrial apoptosis pathway of human renal proximal tubular cells (HK-2 cells). METHODS: Cultured HK-2 cells were inoculated on 6-well plates, according to stochastic tables law, and they were divided into normal serum-treated group (NSTG) , post-asphyxial serum treatment group (PSTG), and post-asphyxial serum and diazoxide treatment group (PSDTG). The serum from neonates 24 hours after asphyxia in a dilution of 20% (volume fraction) was used for challenge. Diazoxide in a final concentration of 100 mol/L, was used for intervention. The expression of caspase-3 was detected by immunohistochemical method. The translocation rate of Omi/HtrA2 and mitochondria membrane potential were determined by indirect immunofluorescence and confocal microscopy. RESULTS: Compared with that of NSTG, the expression of caspase-3 absorbance (A) value of HK-2 cells in PSTG was significantly increased (25.19 + or - 3.33 vs. 13.63 + or - 1.89, P<0.01), the translocation rate of Omi/HtrA2 of HK-2 cells in PSTG was significantly increased [(56.01 + or - 5.30)% vs.(37.59 + or - 5.60)%, P<0.01], mitochondrial membrane red/green fluorescence intensity ratio was decreased significantly (0.79 + or - 1.42 vs. 1.82 + or - 0.23, P<0.01). Compared with the PSTG, the expression value of caspase-3 of HK-2 cells in PSDTG was significantly decreased (20.17 + or - 2.19), the translocation rate of Omi/HtrA2 of HK-2 cells in PSDTG was significantly decreased [(46.91 + or - 2.70)%], and mitochondrial membrane red/green fluorescence intensity ratio increased significantly (1.47 + or - 0.14), but did not recover to the same degree as that of the NSTG (all P<0.01). CONCLUSION: The diazoxide may reduce the expression of caspase-3, intracellular translocation of Omi/HtrA2, and stability of mitochondrial membrane potential, thereby significantly alleviates HK-2 cells injury induced by post-asphyxial-serum of neonate.


Asunto(s)
Apoptosis , Asfixia Neonatal/sangre , Diazóxido/farmacología , Células Epiteliales/patología , Mitocondrias/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3/metabolismo , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Recién Nacido , Túbulos Renales Proximales/citología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Suero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...