Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Biochem Biophys ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38717641

RESUMEN

Ovarian cancer (OC) ranks as the fifth leading cause of cancer-related death in women. The main contributors to the poor prognosis of ovarian cancer are the high rates of recurrence and metastasis. Studies have indicated a crucial role for hepatitis B virus X Ag-Transactivated Protein 8 (XTP8), a protein containing the DEP domain, in various cellular processes, including cell growth, movement, and differentiation, across several types of cancers. However, the role of XTP8 in ovarian cancer remains unclear. We observed elevated expression of XTP8 in ovarian cancer. Silencing XTP8 inhibited cell proliferation, promoted apoptosis, and yielded contrasting results in cells overexpressing XTP8. Furthermore, XTP8 facilitated ovarian cancer invasion and migration, triggering epithelial-mesenchymal transition (EMT). Mechanistically, XTP8 silencing led to reduced phosphorylation levels of AKT, increased p-AMPK levels, and decreased p-mTOR levels, while XTP8 overexpression exerted the opposite effects. Additionally, the activation of p-AMPK rescued the promoting effect of XTP8 on EMT in ovarian cancer cell lines, indicating that XTP8 acts as an oncogene by modulating the AKT/AMPK/mTOR pathway. Through transcriptome sequencing to identify downstream targets of XTP8, we found that XTP8 influences the expression of Caldesmon (CALD1) at both transcriptional and translational levels. CALD1 can be considered a downstream target of XTP8. The collaborative action of XTP8 and CALD1 activates the AKT/AMPK/mTOR pathway, regulating EMT to promote ovarian cancer progression. Inhibiting this signaling axis might represent a potential therapeutic target for ovarian cancer.

2.
J Med Virol ; 96(4): e29510, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38573018

RESUMEN

Hepatitis B virus (HBV) infection poses a significant burden on global public health. Unfortunately, current treatments cannot fully alleviate this burden as they have limited effect on the transcriptional activity of the tenacious covalently closed circular DNA (cccDNA) responsible for viral persistence. Consequently, the HBV life cycle should be further investigated to develop new anti-HBV pharmaceutical targets. Our previous study discovered that the host gene TMEM203 hinders HBV replication by participating in calcium ion regulation. The involvement of intracellular calcium in HBV replication has also been confirmed. In this study, we found that transient receptor potential vanilloid 4 (TRPV4) notably enhances HBV reproduction by investigating the effects of several calcium ion-related molecules on HBV replication. The in-depth study showed that TRPV4 promotes hepatitis B core/capsid protein (HBc) protein stability through the ubiquitination pathway and then promotes the nucleocapsid assembly. HBc binds to cccDNA and reduces the nucleosome spacing of the cccDNA-histones complex, which may regulate HBV transcription by altering the nucleosome arrangement of the HBV genome. Moreover, our results showed that TRPV4 promotes cccDNA-dependent transcription by accelerating the methylation modification of H3K4. In conclusion, TRPV4 could interact with HBV core protein and regulate HBV during transcription and replication. These data suggest that TRPV4 exerts multifaceted HBV-related synergistic factors and may serve as a therapeutic target for CHB.


Asunto(s)
Antineoplásicos , Hepatitis B , Humanos , Ubiquitina , Cápside , Proteínas de la Cápside , Virus de la Hepatitis B/genética , Canales Catiónicos TRPV/genética , Calcio , Nucleosomas , Metilación , Proteínas de la Membrana
3.
Virus Res ; 341: 199326, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38253259

RESUMEN

BACKGROUND: PreS1-binding protein (PreS1BP), recognized as a nucleolar protein and tumor suppressor, influences the replication of various viruses, including vesicular stomatitis virus (VSV) and herpes simplex virus type 1 (HSV-1). Its role in hepatitis B virus (HBV) replication and the underlying mechanisms, however, remain elusive. METHODS: We investigated PreS1BP expression levels in an HBV-replicating cell and animal model and analyzed the impact of its overexpression on viral replication metrics. HBV DNA, covalently closed circular DNA (cccDNA), hepatitis B surface antigen (HBsAg), hepatitis B core antigen (HBcAg), and HBV RNA levels were assessed in HBV-expressing stable cell lines under varying PreS1BP conditions. Furthermore, co-immunoprecipitation and ubiquitination assays were used to detect PreS1BP- hepatitis B virus X protein (HBx) interactions and HBx stability modulated by PreS1BP. RESULTS: Our study revealed a marked decrease in PreS1BP expression in the presence of active HBV replication. Functional assays showed that PreS1BP overexpression significantly inhibited HBV replication and transcription, evidenced by the reduction in HBV DNA, cccDNA, HBsAg, HBcAg, and HBV RNA levels. At the molecular level, PreS1BP facilitated the degradation of HBx in a dose-dependent fashion, whereas siRNA-mediated knockdown of PreS1BP led to an increase in HBx levels. Subsequent investigations uncovered that PreS1BP accelerated HBx protein degradation via K63-linked ubiquitination in a ubiquitin-proteasome system-dependent manner. Co-immunoprecipitation assays further established that PreS1BP enhances the recruitment of the proteasome 20S subunit alpha 3 (PSMA3) for interaction with HBx, thereby fostering its degradation. CONCLUSIONS: These findings unveil a previously unidentified mechanism wherein PreS1BP mediates HBx protein degradation through the ubiquitin-proteasome system, consequentially inhibiting HBV replication. This insight positions PreS1BP as a promising therapeutic target for future HBV interventions. Further studies are warranted to explore the clinical applicability of modulating PreS1BP in HBV therapy.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Animales , Humanos , Virus de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/metabolismo , Proteolisis , Antígenos del Núcleo de la Hepatitis B/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Hep G2 , Proteínas Reguladoras y Accesorias Virales/genética , ADN Circular/metabolismo , Replicación Viral/genética , ARN/metabolismo , Ubiquitinas/genética
4.
Front Pediatr ; 11: 1257491, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37800010

RESUMEN

Neonatal gastric perforation (NGP) is a rare, but life-threatening condition that can lead to serious conditions, such as capillary leak syndrome (CLS). Here, we present the case of a preterm male infant with NGP complicated by CLS after stomach repair. The patient was born at 33 2/7 weeks, weighed 1,770 g, and was diagnosed with respiratory distress syndrome. On the fourth day of life, the patient presented with distention and an unstable cardiovascular system. Routine blood tests revealed a white blood cell count of 2.4 × 109/L. Chest and abdominal radiography revealed a pneumoperitoneum, suggesting a gastrointestinal perforation. The patient was urgently transferred to a tertiary hospital for exploratory laparotomy, where a 2 cm diameter perforation was discovered in the stomach wall and subsequently repaired. Pathological findings indicated the absence of a muscular layer in the stomach wall. The patient unexpectedly developed CLS postoperatively, leading to multiorgan dysfunction and eventual death. The underlying pathological mechanism of NGP-induced CLS may be related to severe chemical peritonitis, sepsis, endothelial glycocalyx dysfunction, enhanced systemic inflammation, and translocation of the gut microbiota, causing endothelial hyperpermeability. Notablely, abdominal surgery itself can be a significant triggering factor for CLS occurrence. Complications of NGP and CLS are extremely dangerous. Investigating the mechanism by which NGP triggers CLS could potentially improve the prognosis. Conservative treatment for pneumoperitoneum secondary to gastric perforation may be a reasonable option, especially when the condition of the patient is unstable.

5.
World J Gastroenterol ; 29(18): 2798-2817, 2023 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-37274069

RESUMEN

BACKGROUND: Hepatic fibrosis is a serious condition, and the development of hepatic fibrosis can lead to a series of complications. However, the pathogenesis of hepatic fibrosis remains unclear, and effective therapy options are still lacking. Our group identified hepatitis C virus nonstructural protein 3-transactivated protein 1 (NS3TP1) by suppressive subtractive hybridization and bioinformatics analysis, but its role in diseases including hepatic fibrosis remains undefined. Therefore, additional studies on the function of NS3TP1 in hepatic fibrosis are urgently needed to provide new targets for treatment. AIM: To elucidate the mechanism of NS3TP1 in hepatic fibrosis and the regulatory effects of calcitriol on NS3TP1. METHODS: Twenty-four male C57BL/6 mice were randomized and separated into three groups, comprising the normal, fibrosis, and calcitriol treatment groups, and liver fibrosis was modeled by carbon tetrachloride (CCl4). To evaluate the level of hepatic fibrosis in every group, serological and pathological examinations of the liver were conducted. TGF-ß1 was administered to boost the in vitro cultivation of LX-2 cells. NS3TP1, α-smooth muscle actin (α-SMA), collagen I, and collagen III in every group were examined using a Western blot and real-time quantitative polymerase chain reaction. The activity of the transforming growth factor beta 1 (TGFß1)/Smad3 and NF-κB signaling pathways in each group of cells transfected with pcDNA-NS3TP1 or siRNA-NS3TP1 was detected. The statistical analysis of the data was performed using the Student's t test. RESULTS: NS3TP1 promoted the activation, proliferation, and differentiation of hepatic stellate cells (HSCs) and enhanced hepatic fibrosis via the TGFß1/Smad3 and NF-κB signaling pathways, as evidenced by the presence of α-SMA, collagen I, collagen III, p-smad3, and p-p65 in LX-2 cells, which were upregulated after NS3TP1 overexpression and downregulated after NS3TP1 interference. The proliferation of HSCs was lowered after NS3TP1 interference and elevated after NS3TP1 overexpression, as shown by the luciferase assay. NS3TP1 inhibited the apoptosis of HSCs. Moreover, both Smad3 and p65 could bind to NS3TP1, and p65 increased the promoter activity of NS3TP1, while NS3TP1 increased the promoter activity of TGFß1 receptor I, as indicated by coimmunoprecipitation and luciferase assay results. Both in vivo and in vitro, treatment with calcitriol dramatically reduced the expression of NS3TP1. Calcitriol therapy-controlled HSCs activation, proliferation, and differentiation and substantially suppressed CCl4-induced hepatic fibrosis in mice. Furthermore, calcitriol modulated the activities of the above signaling pathways via downregulation of NS3TP1. CONCLUSION: Our results suggest that calcitriol may be employed as an adjuvant therapy for hepatic fibrosis and that NS3TP1 is a unique, prospective therapeutic target in hepatic fibrosis.


Asunto(s)
Calcitriol , FN-kappa B , Proteína smad3 , Factor de Crecimiento Transformador beta1 , Proteínas no Estructurales Virales , Animales , Masculino , Ratones , Calcitriol/farmacología , Calcitriol/uso terapéutico , Tetracloruro de Carbono/toxicidad , Colágeno Tipo I/metabolismo , Hepacivirus/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/prevención & control , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteína smad3/metabolismo
6.
J Therm Biol ; 109: 103306, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36195403

RESUMEN

Brown adipose tissue is a thermogenic organ, which consumes chemical energy as heat to protect animals from low temperature and metabolic diseases. However, the role and mechanism of the new factor that up-regulates the heat-generating capacity of brown adipose tissue is still unclear. Here, we found that hepatitis C virus core binding protein 6 (HCBP6), as a key regulator gene in the homeostasis of liver lipid metabolism, is an important enhancer for activating brown fat to ensure thermogenesis. HCBP6 upregulates the expression of UCP1 and increases the number of mitochondria in brown adipocytes. In the BAT of HCBP6-knockout mice induced by a high-fat diet, UCP1 and BAT activity-related genes Pgc1α, Cidea and oxidation phosphorylation-related genes (OXPHOS) were significantly reduced. In addition, the transcriptomics results show that the loss of HCBP6 caused disorder of the metabolic pathway, the expression of brown adipocyte development genes was significantly reduced, and the expression of most BAT cytokine genes was reduced. In conclusion, HCBP6 increased ucp1-dependent thermogenesis in BAT and improved liver lipid metabolism, possibly by enhancing the activity of brown fat and changing the expression of BAT cytokine genes.


Asunto(s)
Tejido Adiposo Pardo , Termogénesis , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Citocinas/genética , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
7.
Sensors (Basel) ; 21(9)2021 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-34065067

RESUMEN

This study innovatively proposes a feature fusion technique to determine fatty acid content during rice storage. Firstly, a self-developed olfactory visualization sensor was used to capture the odor information of rice samples at different storage periods and a portable spectroscopy system was employed to collect the near-infrared (NIR) spectra during rice storage. Then, principal component analysis (PCA) was performed on the pre-processed olfactory visualization sensor data and the NIR spectra, and the number of the best principal components (PCs) based on the single technique model was optimized during the backpropagation neural network (BPNN) modeling. Finally, the optimal PCs were fused at the feature level, and a BPNN detection model based on the fusion feature was established to achieve rapid measurement of fatty acid content during rice storage. The experimental results showed that the best BPNN model based on the fusion feature had a good predictive performance where the correlation coefficient (RP) was 0.9265, and the root mean square error (RMSEP) was 1.1005 mg/100 g. The overall results demonstrate that the detection accuracy and generalization performance of the feature fusion model are an improvement on the single-technique data model; and the results of this study can provide a new technical method for high-precision monitoring of grain storage quality.


Asunto(s)
Oryza , Algoritmos , Ácidos Grasos , Análisis de los Mínimos Cuadrados , Redes Neurales de la Computación , Espectroscopía Infrarroja Corta
8.
Life Sci ; 265: 118848, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33278383

RESUMEN

AIMS: To investigated the effect of S6K1 on the replication and transcription of HBV DNA using multiple cell models. MAIN METHODS: The pgRNA, total HBV RNA and HBV DNA level were detected by Real-time PCR. The HBcAg expression by Western blot and the activity of four HBV promoters, such as preS1, preS2/S, core, and X promoters by using dual luciferase reporter assay. Moreover, we determined S6K1 interacted with HBcAg in both cytoplasm and nucleus through Immunofluorescence, co-immunoprecipitation (CoIP) and Western blot. KEY FINDINGS: S6K1 inhibited HBV DNA replication and cccDNA-dependent transcription in HBV-expressing stable cell lines. The mechanistic study revealed that S6K1 suppressed HBV DNA replication by inhibiting AMPK-ULK1 autophagy pathway, and the nuclear S6K1 suppressed HBV cccDNA-dependent transcription by inhibiting the acetylation modification of H3K27. In addition, HBV capsid protein (HBcAg) suppressed the phosphorylation level of S6K1Thr389 by interacting with S6K1, indicating a viral antagonism of S6K1-mediated antiviral mechanism. SIGNIFICANCE: The p70 ribosomal S6 kinase (S6K1) is a serine/threonine protein kinase, and it plays a significant role in different cellular processes. It has been previously reported that S6K1 affects hepatitis B virus (HBV) replication but the underlying mechanism remains unclear. In this study, our data suggested that the activation of S6K1 restricts HBV replication through inhibiting AMPK-ULK1 autophagy pathway and H3K27 acetylation. These findings indicated that S6K1 might be a potential therapeutic target for HBV infection.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Virus de la Hepatitis B/fisiología , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Replicación Viral/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Acetilación , Autofagia/fisiología , Línea Celular Tumoral , Células Hep G2 , Virus de la Hepatitis B/genética , Humanos , Transducción de Señal/fisiología , Replicación Viral/genética
9.
J Med Virol ; 92(11): 2804-2812, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32542750

RESUMEN

A pandemic of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection broke out all over the world; however, epidemiological data and viral shedding in pediatric patients are limited. We conducted a retrospective, multicenter study, and followed-up with all children from the families with SARS-CoV-2 infected members in Zhejiang Province, China. All infections were confirmed by testing the SARS-CoV-2 RNA with real-time reverse transcription PCR method, and epidemiological data between children and adults in the same families were compared. Effect of antiviral therapy was evaluated observationally and fecal-viral excretion times among groups with different antiviral regiments were compared with Kaplan-Meier plot. By 29 February 2020, 1298 cases from 883 families were confirmed with SARS-CoV-2 infection and 314 of which were families with children. Incidence of infection in child close contacts was significantly lower than that in adult contacts (13.2% vs 21.2%). The mean age of 43 pediatric cases was 8.2 years and mean incubation period was 9.1 days. Forty (93.0%) were family clustering. Thirty-three children had coronavirus disease 2019 (20 pneumonia) with mild symptoms and 10 were asymptomatic. Fecal SARS-CoV-2 RNA detection was positive in 91.4% (32/35) cases and some children had viral excretion time over 70 days. Viral clearance time was not different among the groups treated with different antiviral regiments. No subsequent infection was observed in family contacts of fecal-viral-excreting children. Children have lower susceptibility of SARS-CoV-2 infection, longer incubation, and fecal-viral excretion time. Positive results of fecal SARS-CoV-2 RNA detection were not used as indication for hospitalization or quarantine.


Asunto(s)
COVID-19/epidemiología , Heces/virología , SARS-CoV-2/fisiología , Esparcimiento de Virus , Adolescente , Antivirales/uso terapéutico , COVID-19/transmisión , Portador Sano/epidemiología , Portador Sano/virología , Niño , Preescolar , China/epidemiología , Familia , Femenino , Hospitalización , Humanos , Incidencia , Lactante , Masculino , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2/patogenicidad
10.
Biomed Pharmacother ; 129: 110347, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32535386

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD), which often accompanied by metabolic syndrome, such as obesity, diabetes and dyslipidemia, has become a global health problem. Our previous results show that HCV core protein binding protein 6 (HCBP6) could maintain the triglyceride homeostasis in liver cells. However, the role of HCBP6 in NAFLD and its associated metabolic disorders remains incompletely understood. METHODS: Hepatic HCBP6 expression was determined by qRT-PCR, Western blot and immunohistochemistry analysis. HCBP6 knockout (HCBP6-KO) mice were constructed and fed a high-fat diet (HFD) to induce NAFLD. The effects of HCBP6 on glucose and lipid metabolism were measured by HE staining, qRT-PCR, Western blot and GTT. Wild-type and HCBP6-KO mice kept on a HFD were treated with ginsenosides Rh2, and HE staining and GTT were used to study the function of Rh2 in metabolism disorders. RESULTS: HCBP6 is reduced in HFD-fed mice. HCBP6 deficiency increased the body weight, aggravated fatty liver and deteriorated lipid homeostasis as well as glucose homeostasis in HFD-induced mouse model of NAFLD. Moreover, HCBP6-KO mice failed to maintain body temperature upon cold challenge. Mechanistically, HCBP6 could regulate lipolysis and fatty acid oxidation via activation of AMKP in vivo. In addition, HCBP6 expression was upregulated by ginsenosides Rh2. Accordingly, ginsenosides Rh2 administrations improved HFD-induced fatty liver and glucose tolerance. CONCLUSIONS: These findings indicated that HCBP6 is essential in maintaining lipid and glucose homeostasis and body temperature. HCBP6 augmented by ginsenosides Rh2 may be a promising therapeutic strategy for the treatment of metabolic disorders in NAFLD mice.


Asunto(s)
Proteínas Relacionadas con la Autofagia/deficiencia , Glucemia/metabolismo , Ácidos Grasos/metabolismo , Lipólisis , Hígado/metabolismo , Síndrome Metabólico/metabolismo , Proteínas Mitocondriales/deficiencia , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/genética , Glucemia/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ginsenósidos/farmacología , Células Hep G2 , Humanos , Insulina/sangre , Interleucina-6/metabolismo , Lipólisis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Factor de Necrosis Tumoral alfa/metabolismo
11.
Cancer Sci ; 111(3): 994-1007, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31746531

RESUMEN

The effect of hepatitis C virus p7 trans-regulated protein 3 (P7TP3) in the development of hepatocellular carcinoma (HCC) is still unknown. The present study aimed to investigate the role and mechanism of P7TP3 in HCC. P7TP3 was significantly decreased in HCC tissues when compared with corresponding liver tissues immediately around the tumor (LAT) from seven HCC patients. Fewer and smaller colonies originated from HepG2-P7TP3 cells when compared to HepG2-NC cells. Overexpression of P7TP3 in HepG2 cells significantly repressed the growth of HCC xenografts in nude mice. Furthermore, wound-healing tests, Transwell assays, Matrigel Transwell assays, adhesion assays, CCK-8 assays, flow cytometry and western blotting analysis showed that P7TP3 protein expression inhibited migration, invasion, adhesion, proliferation and cell cycle progression in HCC cell lines. Moreover, P7TP3 suppressed the activity of the Wnt/ß-catenin signaling pathway, and was restored by Wnt3a, which is an activator of the Wnt/ß-catenin signaling pathway. Consistently, ß-catenin was highly expressed by P7TP3 silencing, and restored by XAV939, an inhibitor of the Wnt/ß-catenin signaling pathway. Finally, microRNA (miR)-182-5p suppressed the expression of target gene P7TP3 by directly interacting with the 3'-UTR region. Taken together, P7TP3, the direct target gene of miR-182-5p, inhibited HCC by regulating migration, invasion, adhesion, proliferation and cell cycle progression of liver cancer cell through the Wnt/ß-catenin signaling pathway. These findings provide strong evidence that P7TP3 functions as a new promising tumor suppressor in HCC.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Hepáticas/genética , Transducción de Señal/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , Regiones no Traducidas 3'/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , MicroARNs/genética
12.
Hepatol Int ; 14(1): 145-160, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31758498

RESUMEN

BACKGROUND: This study aimed to investigate the roles and mechanisms of tenofovir alafenamide fumarate (TAF)/tenofovir disoproxil fumarate (TDF) in treating liver fibrosis. METHODS: The effects of TAF/TDF on carbon tetrachloride (CCl4)-induced liver fibrosis in C57BL/6 wild-type or nonstructural protein 5A transactivated protein 9 (NS5ATP9) knockout mice were studied. The differentiation, activation, and proliferation of LX-2 cells after TAF/TDF treatment were tested in vitro. The expression of NS5ATP9 and activities of transforming growth factor-ß1 (TGFß1)/Sekelsky mothers against decapentaplegic homolog 3 (Smad3) and NF-κB/NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome signaling pathways were detected in TAF/TDF-treated mice and LX-2 cells. The genes related to extracellular matrix accumulation were detected in vivo and in vitro after NS5ATP9 silencing or knockout. RESULTS: TAF/TDF significantly inhibited CCl4-induced liver fibrosis in mice, and regulated the differentiation, activation, and proliferation of hepatic stellate cells (HSCs). Furthermore, TAF/TDF suppressed the activities of TGFß1/Smad3 and NF-κB/NLRP3 inflammasome signaling pathways in vivo and in vitro. NS5ATP9 inhibited liver fibrosis through TGFß1/Smad3 and NF-κB signaling pathways. TAF/TDF upregulated the expression of NS5ATP9 in vivo and in vitro. Finally, TAF/TDF could only show marginal therapeutic effects when NS5ATP9 was silenced and knocked out in vivo and in vitro. CONCLUSIONS: TAF/TDF prevented progression and promoted reversion of liver fibrosis through assembling TGFß1/Smad3 and NF-κB/NLRP3 inflammasome signaling pathways via upregulating the expression of NS5ATP9. TAF/TDF also regulated the differentiation, activation, and proliferation of HSCs. The findings provided strong evidence for the role of TAF/TDF as a new promising therapeutic strategy in liver fibrosis.


Asunto(s)
Adenina/análogos & derivados , Antivirales/farmacología , Cirrosis Hepática Experimental/metabolismo , Tenofovir/farmacología , Adenina/farmacología , Alanina , Animales , Tetracloruro de Carbono , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Cirrosis Hepática Experimental/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
Biochem Biophys Res Commun ; 515(3): 455-461, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31164201

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common cancer in the world and the main cause of cancer death. Chronic hepatitis B virus (HBV) infection is the major cause of HCC. HBx, as a transactivator, plays an important role in the occurrence and development process of HCC leading by HBV infection. XTP8, related to HBx, however, there are no studies on the function of XTP8 in HCC. In our research, we demonstrated that XTP8 was significantly up-regulated in HCC tissues compared with non-cancerous tissues in Oncomine, TCGA and GEO database. Moreover, Kaplan-Meier Plotter analysis indicated that patients with higher XTP8 expression had significantly lower overall survival. Our immunohistochemical results suggested that XTP8 protein expression in HCC tissues was dramatically higher compared with control normal tissues. In vivo xenograft experiments on nude mice, the overexpression of XTP8 promoted the tumorigenic ability of HepG2 cells. In HepG2 and Huh7 cells, XTP8 upregulated FOXM1 expression to promote cell proliferation and inhibited cell apoptosis. FOXM1 knockdown reduced promoter activity of XTP8 to downregulate XTP8 expression. Thiostrepton, an inhibitor of FOXM1, decreased XTP8 expression. Therefore, our study demonstrates that XTP8 is a valuable prognostic predictor for HCC and there is a novel positive regulatory feedback loop between XTP8 and FOXM1 promoting the development of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Retroalimentación Fisiológica , Proteína Forkhead Box M1/genética , Proteínas Activadoras de GTPasa/metabolismo , Neoplasias Hepáticas/genética , Oncogenes , Animales , Apoptosis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Proteína Forkhead Box M1/metabolismo , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos
14.
BMC Pediatr ; 19(1): 181, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31167650

RESUMEN

BACKGROUND: Invasive S. pyogenes diseases are uncommon, serious infections with high case fatality rates (CFR). There are few publications on this subject in the field of pediatrics. This study aimed at characterizing clinical and laboratory aspects of this disease in Chinese children. PATIENTS AND METHODS: A retrospective study was conducted and pediatric in-patients with S. pyogenes infection identified by cultures from normally sterile sites were included, who were diagnosed and treated in 9 tertiary hospitals during 2010-2017. RESULTS: A total of 66 cases were identified, in which 37 (56.1%) were male. The median age of these patients, including 11 neonates, was 3.0 y. Fifty-nine (89.4%) isolates were determined from blood. Fever was the major symptom (60/66, 90.9%) and sepsis was the most frequent presentation (64/66, 97.0%, including 42.4% with skin or soft tissue infections and 25.8% with pneumonia. The mean duration of the chief complaint was (3.8 ± 3.2) d. Only 18 (27.3%) patients had been given antibiotics prior to the hospitalization. Among all patients, 15 (22.7%) developed streptococcal toxin shock syndrome (STSS). No S. pyogenes strain was resistant to penicillin, ceftriaxone, or vancomycin, while 88.9% (56/63) and 81.4% (48/59) of the tested isolates were resistant to clindamycin and erythromycin respectively. Most of the patients were treated with ß-lactams antibiotics and 36.4% had been treated with meropenem or imipenem. Thirteen (19.7%) cases died from infection, in which 9 (13.6%) had complication with STSS. CONCLUSIONS: Invasive S. pyogenes infections often developed from skin or soft tissue infection and STSS was the main cause of death in Chinese children. Ongoing surveillance is required to gain a greater understanding of this disease.


Asunto(s)
Infecciones Estreptocócicas/diagnóstico , Streptococcus pyogenes/aislamiento & purificación , Ceftriaxona/uso terapéutico , Preescolar , China , Clindamicina/uso terapéutico , Farmacorresistencia Bacteriana , Eritromicina/uso terapéutico , Femenino , Fiebre/microbiología , Humanos , Lactante , Recién Nacido , Masculino , Penicilinas/uso terapéutico , Neumonía Neumocócica/microbiología , Estudios Retrospectivos , Sepsis/microbiología , Choque Séptico/microbiología , Enfermedades Cutáneas Bacterianas/microbiología , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/efectos de los fármacos , Centros de Atención Terciaria/estadística & datos numéricos , Vancomicina/uso terapéutico
15.
Biochem Biophys Res Commun ; 501(1): 300-306, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29730292

RESUMEN

PURPOSE: To investigate the effect of gestational diabetes mellitus (GDM) on the expression and methylation of PGC-1α and PDX1 in placenta and their effects on fetal glucose metabolism. METHODS: 20 cases of full-term placenta without pregnancy complications and umbilical cord abnormalities and 20 cases of GDM group were collected. DNA and RNA were isolated from samples of tissue collected from the fetal side of the placenta immediately after delivery. DNA methylation was quantified at 7 CpG sites within the PGC-1α and PDX1 genes using PCR amplification of bisulfite treated DNA and subsequent DNA sequencing. PGC-1α and PDX1 mRNA levels were measured by reverse transcription-quantitative PCR (RT-qPCR). Meanwhile, the placental insulin, blood glucose and HbA1c levels were determined. RESULTS: The fetus birth weight and placental weight in GDM group were significantly higher than those in control group (P < 0.05). Insulin, HbA1c and blood glucose levels in GDM group were significantly higher than those in control group (P < 0.01). Insulin content was positively correlated with newborn birth weight and placental weight while HbA1c and blood glucose were positively correlated with insulin concentration (r = 0.92, P < 0.01, r = 0.85, P < 0.01). The levels of PGC-1α and PDX1 mRNA were lower in the GDM group compared to the control group. The methylation level of PGC-1α gene was higher in the GDM group compared to the control group (P < 0.05). Blood glucose was negatively correlated with the expression of PGC-1α and PDX1 mRNA in the placenta (r = -0.42, P < 0.01, r = -0.49, P < 0.01). CONCLUSION: The changes of epigenetic modification of PGC-1α gene in pregnant women with gestational diabetes mellitus may be a mechanism of abnormal glucose metabolism in offspring.


Asunto(s)
Glucemia/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Feto/metabolismo , Proteínas de Homeodominio/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Transactivadores/genética , Adulto , Estudios de Casos y Controles , Islas de CpG , Metilación de ADN , Diabetes Gestacional/sangre , Epigénesis Genética , Femenino , Sangre Fetal/metabolismo , Hemoglobina Glucada/metabolismo , Humanos , Recién Nacido , Insulina/sangre , Placenta/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
16.
Mol Ther Nucleic Acids ; 10: 91-102, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499960

RESUMEN

Recent studies have shown the effect of microRNAs on HSC activation and transformation, which is essential for the pathogenesis of liver fibrosis. In our study, we explored the role of miR-185 in liver fibrosis. Plasma miR-185 was detected in hepatitis B virus-related liver fibrosis patients (S2/3, n = 10) by Illumina HiSeq sequencing, and healthy volunteers were selected (n = 8) as the control group. We found that the plasma miR-185 level in fibrosis patients was significantly downregulated. CCl4-induced fibrosis tissues in mouse livers and TGF-ß1-activated HSCs also presented downregulated miR-185 concomitant with an increased expression of RHEB and RICTOR. To explore the correlations, LX-2 cells were transiently transfected with miR-185 mimics. The expression levels of α-SMA, collagen I, and collagen III were decreased as well as RHEB and RICTOR. Inhibition of endogenous miR-185 increased fibrogenic activity. Furthermore, dual-luciferase reporter assays indicated that miR-185 inhibited the expression of RHEB and RICTOR by directly targeting their 3' UTRs. Moreover, silencing RHEB and RICTOR suppressed α-SMA and collagen expression levels. In conclusion, miR-185 prevents liver fibrogenesis by inhibiting HSC activation via inhibition of RHEB and RICTOR. These results provide new insights into the mechanisms behind the anti-fibrotic effect of miR-185.

17.
BMB Rep ; 51(1): 33-38, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29187281

RESUMEN

Sterol regulatory element-binding protein-1c (SREBP1c) plays an important role in triglyceride (TG) homeostasis. Although our previous study showed that hepatitis C virus core-binding protein 6 (HCBP6) regulates SREBP1c expression to maintain intracellular TG homeostasis, the mechanism underlying this regulation is unclear. In the present study, we found that HCBP6 increased intracellular TG levels by upregulating SREBP1c expression. HCBP6 increased SREBP1c transcription by directly binding to the SREBP1c promoter (at the -139- to +359-bp region). Moreover, we observed that HCBP6 interacted with C/EBPß-binding site in the SREBP1c promoter both in vitro and in vivo. These results indicate that HCBP6 upregulates human SREBP1c expression by binding to the C/EBPß-binding site in the SREBP1c promoter. [BMB Reports 2018; 51(1): 33-38].


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Proteínas del Núcleo Viral/metabolismo , Sitios de Unión , Silenciador del Gen , Células Hep G2 , Hepacivirus/metabolismo , Homeostasis , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Activación Transcripcional , Triglicéridos/metabolismo , Regulación hacia Arriba , Proteínas del Núcleo Viral/genética
18.
J Cell Physiol ; 233(12): 9312-9319, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29227529

RESUMEN

This study aimed to investigate the relationship between interleukin-6 (IL-6) and NS5ATP9 in autophagy of liver cancer cells. Autophagy is one of the important regulators of the replication of hepatitis C virus and the survival of tumors. IL-6 is a multifunctional cytokine that plays an important role in autophagy and development of many kinds of tumors. However, the role of IL-6 in autophagy has not been fully explored. A previous study had shown that a novel gene, NS5ATP9, could modulate autophagy. The present study demonstrated that human IL-6 recombinant protein induced autophagy of HepG2 cells. Conversely, autophagy decreased after IL-6 was silenced or neutralized with monoclonal antibody against human IL-6. In addition, NS5ATP9 was upregulated by IL-6 via nuclear factor-kappaB activation, as detected by Western blot. Further studies indicated that the induction of autophagy by IL-6 could be attenuated by silencing NS5ATP9. Interestingly, the expression of NS5ATP9, in turn, resulted in the upregulation of IL-6. In conclusion, IL-6 could induce autophagy by expressing NS5ATP9, while NS5ATP9 upregulated IL-6 levels in turn, which further induced autophagy.


Asunto(s)
Autofagia , Proteínas de Unión al ADN/genética , Interleucina-6/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Autofagia/efectos de los fármacos , Autofagia/genética , Beclina-1/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Células Hep G2 , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , FN-kappa B/metabolismo , Pruebas de Neutralización , Proteínas Recombinantes/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
19.
Tumour Biol ; 39(2): 1010428317694313, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28240051

RESUMEN

Studies have demonstrated that microRNA 185 may be a promising therapeutic target in liver cancer. However, its role in hepatocellular carcinoma is largely unknown. In this study, the proliferation of human HepG2 cells was inhibited by transfection of microRNA 185 mimics. Cell-cycle analysis revealed arrest at the G0/G1 phase. Transfection of HepG2 cells with microRNA 185 mimics significantly induced apoptosis. These data confirmed microRNA 185 as a potent cancer suppressor. We demonstrated that microRNA 185 was a compelling inducer of autophagy, for the first time. When cell autophagy was inhibited by chloroquine or 3-methyladenine, microRNA 185 induced more cell apoptosis. MicroRNA 185 acted as a cancer suppressor by regulating AKT1 expression and phosphorylation. Dual-luciferase reporter assays indicated that microRNA 185 suppressed the expression of target genes including RHEB, RICTOR, and AKT1 by directly interacting with their 3'-untranslated regions. Binding site mutations eliminated microRNA 185 responsiveness. Our findings demonstrate a new role of microRNA 185 as a key regulator of hepatocellular carcinoma via autophagy by dysregulation of AKT1 pathway.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/genética , Autofagia/genética , Materiales Biomiméticos/administración & dosificación , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Ciclo Celular/genética , Procesos de Crecimiento Celular/genética , Terapia Genética/métodos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , MicroARNs/administración & dosificación , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Transfección/métodos
20.
J Cell Biochem ; 118(8): 2463-2473, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28133777

RESUMEN

Liver fibrosis is a reversible wound-healing response to any etiology of chronic hepatic injuries. Activation of hepatic stellate cells (HSCs) is the key event in liver fibrogenesis. Generally, persistent activation and proliferation of HSCs results in liver fibrosis progression, while primary mechanisms of liver fibrosis resolution are apoptosis and reversion to a quiescent phenotype of activated HSCs. NS5ATP13 (HCV NS5A-transactivated protein 13) is involved in nucleologenesis and tumorigenesis, but its role in liver fibrosis and HSC activation remains unclear. This study found that NS5ATP13 was upregulated in both fibrotic liver tissues and activated human HSCs induced by TGF-ß1. Moreover, NS5ATP13 enhanced extracellular matrix (ECM) production and HSC activation, with or without TGF-ß1 treatment, likely involving the TGF-ß1/Smad3 signaling pathway. Additionally, NS5ATP13 boosted HSC proliferation by inhibiting cell apoptosis. Furthermore, HCV NS5A promoted the profibrogenic effect of NS5ATP13 partly through TGF-ß1 and NF-κB p65 (RelA) upregulation. Meanwhile, NS5ATP13 was required for the pro-fibrogenic effect of NF-κB. Moreover, NS5ATP13 and NF-κB phosphorylation as well as HSC activation were reduced by CX-4945, a CK2 specific inhibitor. These findings indicated that NS5ATP13 acts as a profibrogenic factor, providing a potential target for antifibrotic therapies. J. Cell. Biochem. 118: 2463-2473, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Proteínas/metabolismo , Animales , Western Blotting , Línea Celular , Células Estrelladas Hepáticas/efectos de los fármacos , Inmunohistoquímica , Cirrosis Hepática/genética , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , Naftiridinas/farmacología , Fenazinas , Proteínas/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA