Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Mol Med ; 28(6): e18129, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38426936

RESUMEN

ATP citrate lyase (ACLY), as a key enzyme in lipid metabolism, plays an important role in energy metabolism and lipid biosynthesis of a variety of tumours. Many studies have shown that ACLY is highly expressed in various tumours, and its pharmacological or gene inhibition significantly inhibits tumour growth and progression. However, the roles of ACLY in oesophageal squamous cell carcinoma (ESCC) remain unclear. Here, our data showed that ACLY inhibitor significantly attenuated cell proliferation, migration, invasion and lipid synthesis in different ESCC cell lines, whereas the proliferation, migration, invasion and lipid synthesis of ESCC cells were enhanced after ACLY overexpression. Furthermore, ACLY inhibitor dramatically suppressed tumour growth and lipid metabolism in ESCC cells xenografted tumour model, whereas ACLY overexpression displayed the opposite effect. Mechanistically, ACLY protein harboured acetylated modification and interacted with SIRT2 protein in ESCC cells. The SIRT2 inhibitor AGK2 significantly increased the acetylation level of ACLY protein and inhibited the proliferation and migration of ESCC cells, while overexpression of ACLY partially reversed the inhibitory effect of AGK2 on ESCC cells. Overall, these results suggest that targeting the SIRT2/ACLY signalling axis may be a potential therapeutic strategy for ESCC patients.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Carcinoma de Células Escamosas de Esófago/genética , ATP Citrato (pro-S)-Liasa , Sirtuina 2/genética , Sirtuina 2/metabolismo , Proliferación Celular , Neoplasias Esofágicas/metabolismo , Lípidos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
2.
Mol Carcinog ; 63(5): 897-911, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38353358

RESUMEN

Increasing evidence has demonstrated that glutaminase (GLS) as a key mitochondrial enzyme plays a pivotal role in glutaminolysis, which widely participates in glutamine metabolism serving as main energy sources and building blocks for tumor growth. However, the roles and molecular mechanisms of GLS in esophageal squamous cell carcinoma (ESCC) remains unknown. Here, we found that GLS was highly expressed in ESCC tissues and cells. GLS inhibitor CB-839 significantly suppressed cell proliferation, colony formation, migration and invasion of ESCC cells, whereas GLS overexpression displayed the opposite effects. In addition, CB-839 markedly suppressed glucose consumption and lactate production, coupled with the downregulation of glycolysis-related proteins HK2, PFKM, PKM2 and LDHA, whereas GLS overexpression exhibited the adverse results. In vivo animal experiment revealed that CB-839 dramatically suppressed tumor growth, whereas GLS overexpression promoted tumor growth in ESCC cells xenografted nude mice. Mechanistically, GLS was localized in mitochondria of ESCC cells, which interacted with PDK1 protein. CB-839 attenuated the interaction of GLS and PDK1 in ESCC cells by suppressing PDK1 expression, which further evoked the downregulation of p-PDHA1 (s293), however, GLS overexpression markedly enhanced the level of p-PDHA1 (s293). These findings suggest that interaction of GLS with PDK1 accelerates the glycolysis of ESCC cells by inactivating PDH enzyme, and thus targeting GLS may be a novel therapeutic approach for ESCC patients.


Asunto(s)
Bencenoacetamidas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Glutaminasa , Glucólisis , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Tiadiazoles , Animales , Humanos , Ratones , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Glutaminasa/genética , Glutaminasa/metabolismo , Glucólisis/genética , Ratones Desnudos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo
3.
J Med Virol ; 95(2): e28546, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36734063

RESUMEN

Severe fever with thrombocytopenia syndrome (SFTS) is a life-threatening infectious disease caused by the SFTS virus (SFTSV). This study aimed to evaluate the predictive power of C-reactive protein to lymphocyte ratio (CLR) and establish an early-warning model for SFTS mortality. We retrospectively analyzed hospitalized SFTS patients in six clinical centers from May 2011 to 2022. The efficacy of CLR prediction was evaluated by the receiver operating characteristic (ROC) analysis. A nomogram was established and validated. Eight hundred and eighty-two SFTS patients (median age 64 years, 48.5% male) were enrolled in this study, with a mortality rate of 17.8%. The area under the ROC curve (AUC) of CLR was 0.878 (95% confidence interval [CI]: 0.850-0.903, p < 0.001), which demonstrates high predictive strength. The least absolute shrinkage and selection operator regression selected seven potential predictors. Multivariate logistic regression analysis determined three independent risk factors, including CLR, to construct the nomogram. The performance of the nomogram displayed excellent discrimination and calibration, with significant net benefits in clinical uses. CLR is a brand-new predictor for SFTS mortality. The nomogram based on CLR can serve as a convenient tool for physicians to identify critical SFTS cases in clinical practice.


Asunto(s)
Infecciones por Bunyaviridae , Phlebovirus , Síndrome de Trombocitopenia Febril Grave , Humanos , Masculino , Persona de Mediana Edad , Femenino , Proteína C-Reactiva/análisis , Estudios Retrospectivos , Factores de Riesgo , China
4.
Geriatr Nurs ; 46: 98-104, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35653946

RESUMEN

This study evaluated the effects of traditional Chinese medicine based exercise (TCE) on cognitive outcomes for patients with mild cognitive impairment (MCI), and tried to identify the most effective TCE modality. A systematic review and meta-analysis of randomized controlled trials were conducted. Ten studies met the inclusion criteria. TCE interventions were classified into three types: (a) Tai Chi, (b) Baduanjin, and (c) Qigong. The pooled analysis showed that, overall, TCE had beneficial effects on global cognition and memory. Subgroup analysis further demonstrated that Baduanjin had a larger effect size on global cognition than the other TCE modalities. By contrast, Tai Chi had a larger effect size on memory than the other modalities. This study implied that TCE is a promising exercise option for improving cognition in MCI. However, further studies with a more rigorous study design are warranted to support or falsify the findings of the present review.


Asunto(s)
Disfunción Cognitiva , Taichi Chuan , Anciano , Cognición , Disfunción Cognitiva/terapia , Ejercicio Físico , Terapia por Ejercicio , Humanos , Medicina Tradicional China
5.
J Cell Mol Med ; 24(14): 8206-8220, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32536038

RESUMEN

Long non-coding RNAs (lncRNAs) widely participate in ESCC development and progression; however, the prognostic factors and therapeutic strategies implicated in ESCC development and progression remain to be under investigation. The purpose of the current study was to explore whether WDFY3-AS2 may be a potential prognostic factor and investigate its biological functions in ESCC. Here, WDFY3-AS2 was frequently down-regulated in ESCC tissues and cells, and its expression was correlated with TNM stage, lymph node metastasis and poor prognosis of ESCC patients. Moreover, WDFY3-AS2 down-regulation significantly promoted cell proliferation and invasion, whereas WDFY3-AS2 up-regulation markedly suppressed cell proliferation and invasion in ESCC EC9706 and TE1 cells, coupled with EMT phenotype alterations. WDFY3-AS2 functioned as a competing endogenous RNA (ceRNA) for sponging miR-2355-5p, further resulted in the up-regulation of its target gene SOCS2, followed by suppression of JAK2/Stat5 signalling pathway, to suppress ESCC cell proliferation and invasion in EC9706 and TE1 cells. These findings suggest that WDFY3-AS2 may participate in ESCC development and progression, and may be a novel prognostic factor for ESCC patients, and thus targeting WDFY3-AS2/miR-2355-5p/SOCS2 signalling axis may be a novel therapeutic strategy for ESCC patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Relacionadas con la Autofagia/genética , Carcinoma de Células Escamosas de Esófago/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Interferencia de ARN , ARN Largo no Codificante/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Biomarcadores de Tumor , Línea Celular Tumoral , Movimiento Celular/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/mortalidad , Carcinoma de Células Escamosas de Esófago/patología , Técnica del Anticuerpo Fluorescente , Humanos , Janus Quinasa 2 , Modelos Biológicos , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Análisis de Supervivencia
6.
Clin Infect Dis ; 70(9): 1941-1949, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31240319

RESUMEN

BACKGROUND: The extensive geographical distribution and high mortality rate of severe fever with thrombocytopenia syndrome (SFTS) have made it an important threat to public health. Neutrophil extracellular traps (NETs) can be activated by a variety of pathogens and are associated with thrombocytopenia in viral infections. We aimed to identify NET production and its predictive value for disease progression and prognosis in patients with SFTS. METHODS: A prospective study was performed with a multicenter cohort of patients with SFTS (n = 112) to quantify serum NET levels. Three markers of NETs-namely, cell-free DNA (cfDNA), myeloperoxidase-DNA complexes, and lactoferrin-DNA complexes-were measured with PicoGreen double-stranded DNA assays and enzyme-linked immunosorbent assays. Receiver operating characteristic curves and multivariate regression analyses were performed to calculate the predictive value of cfDNA levels. RESULTS: SFTS was characterized by pronounced NET formation. The serum levels of NETs changed dynamically during disease progression, with an inverse pattern of the trends of platelet and neutrophil levels. High cfDNA levels were strongly associated with multiple pathological processes, including coagulopathy, myocardial damage, liver dysfunction, and the development of encephalopathy. A high level of cfDNA (>711.7 ng/mL) at the time of the initial diagnosis predicted severe illness in patients with SFTS (odds ratio, 8.285 [95% confidence interval, 2.049-33.503]; P = .003). CONCLUSIONS: This study has a high degree of clinical impact for identification of cfDNA as a useful predictive biomarker of clinical outcomes of SFTS.


Asunto(s)
Ácidos Nucleicos Libres de Células , Phlebovirus , Síndrome de Trombocitopenia Febril Grave , Trombocitopenia , Humanos , Phlebovirus/genética , Pronóstico , Estudios Prospectivos
7.
BMC Med Genet ; 21(1): 4, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31906877

RESUMEN

BACKGROUND: Chediak-Higashi Syndrome (CHS) is a rare autosomal recessive disease caused by loss of function of the lysosomal trafficking regulator protein. The causative gene LYST/CHS1 was cloned and identified in 1996, which showed significant homology to other species such as bovine and mouse. To date, 74 pathogenic or likely pathogenic mutations had been reported. CASE PRESENTATION: Here we describe a compound heterozygote in LYST gene, which was identified in a 4-year-old female patient. The patient showed skin hypopigmentation, sensitivity to light, mild splenomegaly and reduction of platelets in clinical examination. Giant intracytoplasmic inclusions were observed in the bone marrow examination, suggesting the diagnosis of CHS. Amplicon sequencing was performed to detect pathogenic mutation in LYST gene. The result was confirmed by two-generation pedigree analysis base on sanger sequencing. CONCLUSION: A compound heterozygote in LYST gene, consisting of a missense mutation c.5719A > G and an intron mutation c.4863-4G > A, was identified from the patient by using amplicon sequencing. The missense mutation is reported for the first time. Two-generation pedigree analysis showed these two mutations were inherited from the patient's parents, respectively. Our result demonstrated that amplicon sequencing has great potential for accelerating and improving the diagnosis of rare genetic diseases.


Asunto(s)
Secuencia de Aminoácidos/genética , Síndrome de Chediak-Higashi/genética , Proteínas de Transporte Vesicular/genética , Síndrome de Chediak-Higashi/patología , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Lisosomas/genética , Mutación Missense , Linaje
8.
J Recept Signal Transduct Res ; 39(2): 114-121, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31322033

RESUMEN

Objectives: Mounting evidence has demonstrated that C-Phycocyanin (C-PC) exhibits marked antitumor activity in a wide type of tumors, such as pancreas cancer, breast carcinoma, lung cancer, and colon cancer. The current study aimed to confirm the antitumor efficacy of C-PC in esophageal squamous cell carcinoma (ESCC). Methods: The efficacy of C-PC was evaluated against the proliferation of ESCC cell lines EC9706 and EC1 by CCK-8 kit and in a mice model of ESCC EC9706. Cell cycle and apoptosis were investigated by flow cytometry, and cell invasion was determined via transwell chamber. Protein expression was examined by Western blots. Results: We found that C-PC exhibited anti-proliferation ability in a time-dependent manner and a dose-dependent manner in ESCC EC9706 and EC1 cells. Besides, C-PC markedly arrested cell cycle in the G0/G1 phase, induced cell apoptosis and suppressed cell invasion ability in both EC9706 and EC1 cells (p < .01). Notably, C-PC evoked the elevations of Bax, PARP, and cleaved-caspase-3 protein, but reduced cyclin D1, CDK4, Bcl-2, MMP-2, and MMP-9 expression levels. Further investigation from in vivo experiment revealed that C-PC displayed significant antitumor efficacy in the xenografted EC9706 model. Conclusions: Our data presented herein suggest C-PC exerts antitumor efficacy in ESCC.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Proteínas de Neoplasias/genética , Ficocianina/farmacología , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
9.
Tumour Biol ; 39(5): 1010428317708267, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28488526

RESUMEN

Mounting evidence has demonstrated that Bit1 has been investigated as an etiological factor for certain cancers, including esophageal squamous cell carcinoma reported in our previous study, but data regarding possible roles of Bit1 in esophageal squamous cell carcinoma and esophageal adenocarcinoma remain to be elucidated. The purpose of this study was to examine whether Bit1 can be a novel diagnostic marker for the patients with esophageal squamous cell carcinoma and esophageal adenocarcinoma. The results revealed that Bit1 level in esophageal squamous cell carcinoma was significantly higher than that in esophageal adenocarcinoma tissues ( p < 0.05); notably, Bit1 level in esophageal adenocarcinoma tissues was lower than that in paired normal tissues but no difference was found ( p > 0.05). Bit1 expression patterns were completely in accordance with matrix metalloproteinase 2 and Bcl-2 in esophageal squamous cell carcinoma and esophageal adenocarcinoma. In addition, Bit1, Bcl-2, and matrix metalloproteinase 2 expression patterns in different differentiated esophageal squamous cell carcinoma were higher than those in corresponding normal esophageal tissues. Bit1 expression in poorly differentiated esophageal squamous cell carcinoma was significantly higher than that in normal esophageal tissues ( p < 0.05) but not in moderately and well-differentiated esophageal squamous cell carcinoma. Matrix metalloproteinase 2 expression patterns in poorly and moderately differentiated esophageal squamous cell carcinoma were significantly higher than those in corresponding normal esophageal tissues ( p < 0.01) but not in well-differentiated esophageal squamous cell carcinoma tissue ( p > 0.05). Bcl-2 expression patterns in various differentiated esophageal squamous cell carcinoma were higher than those in corresponding normal esophageal tissues with no statistical differences ( p > 0.05). Importantly, Bit1 expression was positively correlated with both matrix metalloproteinase 2 and Bcl-2 expression in esophageal squamous cell carcinoma and esophageal adenocarcinoma tissues ( p < 0.05). Collectively, these preliminary data support further investigation of Bit1 as an important diagnostic factor for esophageal squamous cell carcinoma and esophageal adenocarcinoma.


Asunto(s)
Adenocarcinoma/genética , Hidrolasas de Éster Carboxílico/biosíntesis , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Metaloproteinasa 2 de la Matriz/biosíntesis , Proteínas Mitocondriales/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Adenocarcinoma/patología , Adulto , Anciano , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Hidrolasas de Éster Carboxílico/genética , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/biosíntesis
10.
Mol Cancer ; 15: 23, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26956728

RESUMEN

BACKGROUND: There is growing evidence that Bit1 exerts different roles in the development and progression of human cancers. Although Bit1 was highly exhibited in ESCC tissues in our previous study, its roles and molecular mechanisms implicated in development and progression of ESCC remain unknown. METHODS: Bit1 protein expression in ESCC cell lines and normal esophageal epithelial cell was detected by Western blotting. Bit1 protein expression mediated by Bit1 shRNA was investigated by Western blotting. MTT, migration assay, invasion experiment, ELISA and Flow cytometry were utilized to determine the effects of Bit1 knockdown on cell proliferation, migration, invasion and apoptosis, respectively. A xenograft model was used to examine in vivo tumourigenicity, and immunohistochemistry and TUNEL were utilized to evaluate the related protein expression and apoptosis. Gene microarray was determined by Agilent SurePrint G3 Human GE 8 × 60 K Microarray, the interaction of Bit1 and FAK proteins were detected by Immunoprecipitation and the key protein expressions of FAK-paxillin pathway were detected by Western blotting. RESULTS: We found Bit1 expression in all human ESCC cell lines tested was significantly higher than that in normal esophageal epithelial cell Het-1A (P < 0.05), in which EC9706 presented the highest Bit1 level. Bit1 protein level was significantly downregulated at day 1 after transfection with specific shRNA against Bit1 (P < 0.05). At days 2 and 3, Bit1 level reached the lowest value after transfection with Bit1 shRNA. Moreover, Bit1 depletion contributed to growth inhibition in vitro and in vivo, reduced cell migration and invasion abilities, and induced cell apoptosis in EC9706 and TE1 cells. More importantly, Bit1 downregulation significantly lowered Bcl-2 and MMP-2 levels in EC9706 xenografted tumor tissues, meanwhile triggered apoptosis after treatment with different doses of Bit1 shRNA. Further gene microarray revealed that 23 genes in Bit1-RNAi group were markedly downregulated, whereas 16 genes were obviously upregulated. Notably, Bit1 intrinsically interacted with FAK protein in EC9706 cells. Moreover, paxillin was downregulated at mRNA and protein levels in Bit1 shRNA group, coupled with the decreases of FAK mRNA and protein expressions. CONCLUSION: Bit1 may be an important regulator in cell growth, apoptosis, migration and invasion of ESCC via targeting FAK-paxillin pathway, and thereby combinative manipulation of Bit1 and FAK-paxillin pathway may be the novel and promising therapeutic targets for the patients with ESCC.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Neoplasias Esofágicas/patología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas Mitocondriales/metabolismo , Paxillin/metabolismo , Animales , Apoptosis , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Carcinoma de Células Escamosas de Esófago , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Desnudos , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Tumour Biol ; 35(1): 519-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23955799

RESUMEN

Overwhelming evidence has demonstrated that Bit1 and AIF as mitochondrial proteins are implicated in the development and progression of a variety of tumors. However, their expressions and biological functions in esophageal squamous cell carcinoma (ESCC) remain to be delineated. In the present study, we found that Bit1, AIF, and Bcl-2 levels in ESCC tissues were significantly higher than those in normal esophageal epithelial tissues and dysplasia tissues (P < 0.05). Stepwise investigation demonstrated that Bit1 and Bcl-2 levels were both tightly associated with lymphatic metastasis and TNM staging (P < 0.05), and the levels of Bit1 mRNA as well as AIF and Bcl-2 proteins were both closely related to tumor differentiation (P < 0.05), but not related to the patients' age and gender (P > 0.05). Importantly, Bit1 mRNA and protein levels in ESCC with lymphatic metastasis and TNM staging in III and IV were markedly higher than that without lymphatic metastasis and TMN staging in I and II. Further analysis showed that expression of Bit1 protein was both positively correlated with expressions of AIF and Bcl-2 proteins (r = 0.408 and 0.405, respectively; P < 0.05). Correctively, our data cited earlier suggest that Bit1 plays pivotal roles in the development and progression of ESCC, and its biological functions in ESCC may be closely associated with AIF and Bcl-2 levels.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Hidrolasas de Éster Carboxílico/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Expresión Génica , Proteínas Mitocondriales/genética , Adulto , Anciano , Factor Inductor de la Apoptosis/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/metabolismo , Metástasis de la Neoplasia , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Zhonghua Yi Xue Za Zhi ; 94(48): 3848-53, 2014 Dec 30.
Artículo en Zh | MEDLINE | ID: mdl-25623321

RESUMEN

OBJECTIVE: To explore the effects of membrane skeleton protein 4.1R on the efficiency of photodynamic therapy (PDT). METHODS: 4.1R gene knockout and wild-type mouse embryonic fibroblasts (MEFs) were incubated with various concentrations of 5-aminolevulinic acid (5-ALA) (0.25, 0.50, 1.00, 1.50 and 2.00 mmol/L), followed by exposure to 450 nm light at a dose of 72, 96, 120, 180, 240 mJ/cm(2). Cell counting kit 8 (CCK-8) assay was used to assess the survival rate after PDT treatment. Laser confocal microscopy was used to observe the location of photo-sensitizer protoporphyrin and fluorescence spectrophotometer for detecting the fluorescent intensity of intracellular protoporphyrin. The protein levels of rate-limiting enzyme of protoporphyrin synthesis, ferrochelatase (FECH) and hydroxymethylbilane synthase (HMBS) were determined by Western blot. RESULTS: Both cell lines were killed after 5-aminolevulinic acid (5-ALA)-PDT and its efficacy was dependent on 5-ALA concentration, incubation duration and light dose. The cell survival rates of 4.1R(-/-) MEF were significantly higher than those of 4.1R(+/+) MEF (46.9% ± 7.1% vs 12.5% ± 2.1%, P < 0.001) after PDT treatment with a light dose of 120 mJ/cm(2) mediated by 5-ALA 1.00 mmol/L. After incubation with 1.00 mmol/L 5-ALA, protoporphyrin was distributed throughout cytoplasm in both cell lines while the fluorescent intensity of 4.1R(+/+) MEF was higher than that of 4.1R(-/-) MEF (124.2 ± 3.5 vs 34.6 ± 3.8, P < 0.001). Western blot showed that no difference of FECH and HMBS protein level was found in two cell lines. CONCLUSIONS: A lack of protein 4.1R may attenuate the intracellular protoporphyrin level and the photo-cytotoxicity of PDT. No cellular change of ALA metabolic activity is found. Protein 4.1R may be involved in the ALA uptake in MEF cells so that the cellular level of protoporphyrin ultimately affects the PDT efficiency.


Asunto(s)
Fotoquimioterapia , Ácido Aminolevulínico , Animales , Línea Celular , Ratones , Proteínas de Microfilamentos , Microscopía Confocal , Fármacos Fotosensibilizantes , Protoporfirinas
13.
Int J Mol Med ; 54(3)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38994756

RESUMEN

Drug resistance is a key factor underlying the failure of tumor chemotherapy. It enhances the stem­like cell properties of cancer cells, tumor metastasis and relapse. Luteolin is a natural flavonoid with strong anti­tumor effects. However, the mechanism(s) by which luteolin protects against paclitaxel (PTX)­resistant cancer cell remains to be elucidated. The inhibitory effect of luteolin on the proliferation of EC1/PTX and EC1 cells was detected by cell counting kit­8 assay. Colony formation and flow cytometry assays were used to assess clonogenic capacity, cell cycle and apoptosis. Wound healing and Transwell invasion tests were used to investigate the effects of luteolin on the migration and invasion of EC1/PTX cells. Western blotting was used to detect the protein levels of EMT­related proteins and stem cell markers after sphere formation. Parental cells and drug­resistant cells were screened by high­throughput sequencing to detect the differential expression of RNA and differential genes. ELISA and western blotting were used to verify the screened PI3K/Akt signaling pathway, key proteins of which were explored by molecular docking. Hematoxylin and eosin staining and TUNEL staining were used to observe tumor xenografts on morphology and apoptosis in nude mice. The present study found that luteolin inhibited tumor resistance (inhibited proliferation, induced cell cycle arrest and apoptosis and hindered migration invasion, EMT and stem cell spherification) in vitro in PTX­resistant esophageal squamous cell carcinoma (ESCC) cells. In addition, luteolin enhanced drug sensitivity and promoted the apoptosis of drug­resistant ESCC cells in combination with PTX. Mechanistically, luteolin may inhibit the PI3K/AKT signaling pathway by binding to the active sites of focal adhesion kinase (FAK), Src and AKT. Notably, luteolin lowered the tumorigenic potential of PTX­resistant ESCC cells but did not show significant toxicity in vivo. Luteolin enhanced drug chemosensitivity by downregulating the FAK/PI3K/AKT pathway in PTX­resistant ESCC and could be a promising agent for the treatment of PTX­resistant ESCC cancers.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Luteolina , Paclitaxel , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Luteolina/farmacología , Paclitaxel/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Línea Celular Tumoral , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/genética , Transducción de Señal/efectos de los fármacos , Ratones , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Ratones Desnudos , Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto , Antineoplásicos Fitogénicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Masculino
14.
Mol Biol Rep ; 40(10): 5723-32, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24057236

RESUMEN

There is growing evidence that inhibitor of growth 4 (ING4) plays a pivotal role in development and progression of multiple different tumors; however, its precise function in gastric carcinoma remains to be elucidated. In the present study, we investigated ING4 level in gastric carcinoma tissues and cells, and preliminarily elucidated the role of ING4 in the proliferation and invasion of gastric carcinoma. The results demonstrated that expressions of ING4 mRNA and protein in gastric carcinoma tissues and cells were significantly lower than those in normal tissues and cells (P < 0.05). ING4 level in gastric carcinoma cells stably expressing ING4 was markedly higher than those in untreated group and empty vector pcDNA3.1 group (P < 0.05). Elevated ING4 level resulted in the inhibition of proliferation and invasion in three of gastric carcinoma cell lines MKN-28, SGC-7901 and MKN-45. Most notably, increased ING4 level evidently evoked the down-regulation of p65, p-IκBα, MMP-9 and uPA proteins and the up-regulation of IκBα protein. Our results presented herein suggest that ING4 level elevation mediated proliferation and invasion inhibition may be tightly associated with the suppression of NF-κB signaling pathway.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Homeodominio/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Neoplasias Gástricas/genética , Transfección , Proteínas Supresoras de Tumor/genética
15.
Can J Physiol Pharmacol ; 91(12): 1112-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24289083

RESUMEN

Roxithromycin is an oral macrolide antibiotic agent that has been repeatedly reported to provoke excessive prolongation of the Q-T interval and torsades de pointes in clinical settings. To investigate the mechanisms underlying the arrhythmogenic side effects of roxithromycin, we studied the molecular mechanisms of roxithromycin on human ether-à-go-go-related gene (hERG) K(+) channels expressed in human embryonic kidney (HEK293) cells. Roxithromycin was found to inhibit wild-type (WT) hERG currents in a concentration-dependent manner with a half-maximum block concentration (IC50) of 55.8 ± 9.1 µmol/L. S6 residue hERG mutants (Y652A and F656C) showed reduced levels of hERG current blockage attributable to roxithromycin. Roxithromycin also inhibited the trafficking of hERG protein to the cell membrane, as confirmed by Western blot analysis and confocal microscopy. These findings indicate that roxithromycin may cause acquired long-QT syndrome via direct inhibition of hERG current and by disruption of hERG protein trafficking. Mutations in drug-binding sites (Y652A or F656C) of the hERG channel were found to attenuate hERG current blockage by roxithromycin, but did not significantly alter the disruption of trafficking.


Asunto(s)
Síndrome de QT Prolongado/tratamiento farmacológico , Potenciales de la Membrana/efectos de los fármacos , Roxitromicina/farmacología , Transactivadores/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Células HEK293 , Humanos , Síndrome de QT Prolongado/metabolismo , Potenciales de la Membrana/genética , Mutación/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Transactivadores/genética , Regulador Transcripcional ERG
16.
J Oncol ; 2023: 2205083, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36733672

RESUMEN

Gastric cancer (GC) is a highly molecular heterogeneous tumor with unfavorable outcomes. The Notch signaling pathway is an important regulator of immune cell differentiation and has been associated with autoimmune disorders, the development of tumors, and immunomodulation caused by tumors. In this study, by developing a gene signature based on genes relevant to the Notch pathway, we could improve our ability to predict the outcome of patients with GC. From the TCGA database, RNA sequencing data of GC tumors and associated normal tissues were obtained. Microarray data were collected from GEO datasets. The Molecular Signature Database (MSigDB) was accessed in order to retrieve sets of human Notch pathway-related genes (NPRGs). The LASSO analysis performed on the TCGA cohort was used to generate a multigene signature based on prognostic NPRGs. In order to validate the gene signature, the GEO cohort was utilized. Using the CIBERSORT method, we were able to determine the amounts of immune cell infiltration in the GC. In this study, a total of 21 differentially expressed NPRGs were obtained between GC specimens and nontumor specimens. The construction of a prognostic prediction model for patients with GC involved the identification and selection of three different NPRGs. According to the appropriate cutoff value, the patients with GC were divided into two groups: those with a low risk and those with a high risk. The time-dependent ROC curves demonstrated that the new model had satisfactory performance when it came to prognostic prediction. Multivariate assays confirmed that the risk score was an independent marker that may be used to predict the outcome of GC. In addition, the generated nomogram demonstrated a high level of predictive usefulness. Moreover, the scores of immunological infiltration of the majority of immune cells were distinctly different between the two groups, and the low-risk group responded to immunotherapy in a significantly greater degree. According to the results of a functional enrichment study of candidate genes, there are multiple pathways and processes associated with cancer. Taken together, a new gene model associated with the Notch pathway may be utilized for the purpose of predicting the prognosis of GC. One potential method of treatment for GC is to focus on NPRGs.

17.
Transl Oncol ; 29: 101626, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36689863

RESUMEN

Increasing evidence has demonstrated that Ctr1 plays a crucial role in the regulation of cisplatin uptake in a variety of tumors. The purpose of this study was to investigate its role in mediating cisplatin sensitivity in ESCC cells. Immunohistochemistry (IHC), In situ hybridization (ISH) and semi-quantitative RT-PCR were used to detect Ctr1 expressions in ESCC tissues. qRT-PCR and Western blot was performed to investigate the levels of Ctr1 mRNA and protein in ESCC cells. CCK-8, Flow cytometry and Transwell chamber assay were carried out to examine cell proliferation, apoptosis, migration and invasion abilities in ESCC cells. We found that ESCC tissues and cells had higher Ctr1 level than normal tissues and Het-1A cell. Ctr1 expression was correlated with histological grade, invasion depth, TNM staging and lymph node metastasis in ESCC patients. Ctr1 depletion reduced the suppressive role of proliferation, migration and invasion as well as the inductive role of cell apoptosis and Caspase-3 activity evoked by cisplatin, whereas Ctr1 upregulation combined with cisplatin exerted the synergistic role in regulation of proliferation, apoptosis, Caspase-3 activity, migration and invasion in ESCC. In conclusion, Ctr1 is implicated in ESCC development and progression and its expression may be a novel predictor for assessment of cisplatin sensitivity in ESCC.

18.
Am J Transl Res ; 15(4): 2426-2442, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37193148

RESUMEN

BACKGROUND: Long intergenic non-protein coding RNA 707 (LINC00707) has been identified as a cancer-associated long non-coding RNA (lncRNA) in a variety of cancers. However, the functions and molecular mechanisms of LINC00707 in esophageal squamous cell carcinoma (ESCC) are still unclear. METHODS: The expression of LINC00707 in esophageal cancer (ESCA) and ESCC tissues was determined by online tools, RNA-sequence (RNA-seq) dataset, and quantitative real time polymerase chain reaction (qRT-PCR). The associations between LINC00707 expression and clinicopathologic features and prognosis were investigated. Furthermore, the expression of LINC00707 in ESCC cell lines was determined by qRT-PCR. Then, using LncACTdb 2.0 database, combined with loss-of-function assay verification, we investigated the biologic role of LINC00707 in ESCC cell growth, apoptosis, invasion, and migration by CCK-8, colony formation, flow cytometry and transwell assays. Finally, western blot was used to evaluate the regulatory effect of LINC00707 on PI3K/Akt signaling pathway. RESULTS: Increased LINC00707 expression was exhibited in ESCC tissues and cell lines. High expression of LINC00707 was positively associated with higher tumor-node-metastasis (TNM) stage and lymph node metastasis. Furthermore, LINC00707 expression was significantly higher in patients who drink alcohol, have lymph node metastasis, and harbor higher tumor stage. In addition, Kaplan-Meier survival analysis and receiver operating characteristic (ROC) curve confirmed the feasibility of LINC00707 as a prognostic signature or diagnostic marker. Functional experiments showed that LINC00707 downregulation suppressed ESCC cell proliferation, and metastasis, and induced ESCC cell apoptosis. Mechanistic investigation demonstrated that LINC00707 activated the PI3K/Akt signaling pathway in ESCC cells. CONCLUSIONS: Our findings suggest LINC00707 functions as an oncogenic lncRNA in ESCC, and imply that LINC00707 may be a promising prognostic biomarker and therapeutic target for ESCC patients.

19.
Am J Transl Res ; 15(3): 2109-2121, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37056805

RESUMEN

Macrophages are the core of the pathophysiology of rheumatoid arthritis (RA). They participate in specific and non-specific immunological responses, have phagocytosis, chemotaxis and immune regulatory functions, and are involved in the onset and progression of RA. In recent years, research on the pathophysiology of RA has focused on the polarization and functions of classically activated M1 and selectively activated M2 macrophage subtypes. M1 macrophages release different proinflammatory cytokines, thus driving the chronic proinflammatory, tissue destruction and pain response in RA. M2 macrophages play an anti-inflammatory role. Because of the important role of monocyte-macrophage in RA, drug research targeting monocyte-macrophage can bring us more hope for treatment of RA. This study reviewed the characteristics, plasticity, molecular activation mechanism and relationship of RA with mononuclear macrophages, as well as the transformative potential of macrophages in developing new therapeutic drugs for clinical practice.

20.
Tumour Biol ; 33(5): 1363-74, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22477709

RESUMEN

Emerging evidence indicates that RhoE as novel member of the Rho GTPases family plays an essential role in carcinogenesis and tumor progression of human various tumors, but the functional significance of RhoE in human esophageal squamous cell carcinoma (ESCC) is still unclear. In the current study, RhoE expression in ESCC tissues and cells was examined, and the biological functions of RhoE in ESCC cells were determined. The results revealed that RhoE expression at mRNA and protein levels was significantly downregulated in ESCC tissues and cell lines (P < 0.05). RhoE expression was tightly associated with differentiation degree, clinical staging, and lymph node metastasis of the patients with ESCC (P < 0.05), but no significant correlations were found between RhoE expression and gender or age of the patients with ESCC (P > 0.05). Additionally, we found that downregulation of RhoE expression in ESCC cells promoted cell proliferation, cell cycle progression, as well as cell invasion in vitro, and inhibited cell apoptosis. Conversely, upregulation of RhoE expression in ESCC cells inhibited cell proliferation, arrested cell cycle at G0/G1 phase, reduced cell invasion, and promoted cell apoptosis. Furthermore, the downregulation of RhoE expression significantly reduced PTEN level and enhanced pAkt level; however, elevation of RhoE expression markedly increased PTEN level and decreased pAkt level. Stepwise investigations demonstrated that overexpression of RhoE in ESCC cells increased the expressions of p27 and bax proteins but decreased the expressions of cyclin D1 and bcl-2 proteins. These data demonstrate that RhoE may play a driving role in the development and progression of ESCC, and targeting the RhoE may be an effective and feasible approach for treatment of ESCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Adulto , Anciano , Apoptosis/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Interferencia de ARN , Proteínas de Unión al GTP rho/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA