Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Int J Surg ; 110(3): 1627-1636, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38116670

RESUMEN

PURPOSE: The aim of this study was to develop a nomogram for predicting the probability of postoperative soiling in patients aged greater than 1 year operated for Hirschsprung disease (HSCR). MATERIALS AND METHODS: The authors retrospectively analyzed HSCR patients with surgical therapy over 1 year of age from January 2000 and December 2019 at our department. Eligible patients were randomly categorized into the training and validation set at a ratio of 7:3. By integrating the least absolute shrinkage and selection operator [LASSO] and multivariable logistic regression analysis, crucial variables were determined for establishment of the nomogram. And, the performance of nomogram was evaluated by C-index, area under the receiver operating characteristic curve, calibration curves, and decision curve analysis. Meanwhile, a validation set was used to further assess the model. RESULTS: This study enrolled 601 cases, and 97 patients suffered from soiling. Three risk factors, including surgical history, length of removed bowel, and surgical procedures were identified as predictive factors for soiling occurrence. The C-index was 0.871 (95% CI: 0.821-0.921) in the training set and 0.878 (95% CI: 0.811-0.945) in the validation set, respectively. And, the AUC was found to be 0.896 (95% CI: 0.855-0.929) in the training set and 0.866 (95% CI: 0.767-0.920) in the validation set. Additionally, the calibration curves displayed a favorable agreement between the nomogram model and actual observations. The decision curve analysis revealed that employing the nomogram to predict the risk of soiling occurrence would be advantageous if the threshold was between 1 and 73% in the training set and 3-69% in the validation set. CONCLUSION: This study represents the first efforts to develop and validate a model capable of predicting the postoperative risk of soiling in patients aged greater than 1 year operated for HSCR. This model may assist clinicians in determining the individual risk of soiling subsequent to HSCR surgery, aiding in personalized patient care and management.


Asunto(s)
Nomogramas , Niño , Humanos , Calibración , Periodo Posoperatorio , Estudios Retrospectivos , Factores de Riesgo , Lactante
2.
Oncogene ; 42(36): 2673-2687, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37537343

RESUMEN

Malate-aspartate shuttle (MAS) is essential for maintaining glycolysis and energy metabolism in tumors, while its regulatory mechanisms in neuroblastoma (NB), the commonest extracranial malignancy during childhood, still remain to be elucidated. Herein, by analyzing multi-omics data, GATA binding protein 2 (GATA2) and its antisense RNA 1 (GATA2-AS1) were identified to suppress MAS during NB progression. Mechanistic studies revealed that GATA2 inhibited the transcription of glutamic-oxaloacetic transaminase 2 (GOT2) and malate dehydrogenase 2 (MDH2). As a long non-coding RNA destabilized by RNA binding motif protein 15-mediated N6-methyladenosine methylation, GATA2-AS1 bound with far upstream element binding protein 3 (FUBP3) to repress its liquid-liquid phase separation and interaction with suppressor of zest 12 (SUZ12), resulting in decrease of SUZ12 activity and epigenetic up-regulation of GATA2 and other tumor suppressors. Rescue experiments revealed that GATA2-AS1 inhibited MAS and NB progression via repressing interaction between FUBP3 and SUZ12. Pre-clinically, administration of lentivirus carrying GATA2-AS1 suppressed MAS, aerobic glycolysis, and aggressive behaviors of NB xenografts. Notably, low GATA2-AS1 or GATA2 expression and high FUBP3, SUZ12, GOT2 or MDH2 levels were linked with unfavorable outcome of NB patients. These findings suggest that GATA2-AS1 inhibits FUBP3 phase separation to repress MAS and NB progression via modulating SUZ12 activity.


Asunto(s)
Neuroblastoma , ARN Largo no Codificante , Humanos , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Malatos/metabolismo , Línea Celular Tumoral , ARN sin Sentido , Neuroblastoma/patología , ARN Largo no Codificante/genética , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Factor de Transcripción GATA2/genética
5.
J Pediatr Urol ; 19(3): 313-319, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36966014

RESUMEN

BACKGROUND: The surgical approach for high-level intra-abdominal testis (IAT) is variable. While most pediatric urologists prefer staged Fowler-Stephens orchiopexy (FSO), Shehata publicized a novel approach-known as staged laparoscopic traction orchiopexy (SLTO) or the Shehata technique-to better manage IATs. OBJECTIVE: This study compares the overall success rates, atrophy rates, retraction rates, and operation times of the two procedures to assist surgeons with developing procedure strategies. METHODS: Databases were searched for relevant literature involving these two approaches, and studies meeting the eligibility criteria were involved; RevMan 5.4 was used to conduct this meta-analysis. The relative risk (RR), weighted mean difference, 95% confidence interval (CI), p-value, publication bias, and heterogeneity were calculated. RESULTS: The Shehata technique demonstrated better performance than staged FSO regarding the overall success and atrophy rate, while the retraction rate and operation time had no statistical difference. CONCLUSIONS: This study revealed that the Shehata technique may be an alternative to staged FSO for managing high-level IATs. Additional high-quality studies regarding the Shehata technique, as well as a long-term follow-up, are required for further and more credible analysis.


Asunto(s)
Cavidad Abdominal , Criptorquidismo , Laparoscopía , Masculino , Niño , Humanos , Orquidopexia/métodos , Criptorquidismo/cirugía , Testículo/cirugía , Cavidad Abdominal/cirugía , Atrofia , Laparoscopía/métodos
6.
Front Pediatr ; 10: 834465, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35311041

RESUMEN

Background: Pneumovesicoscopic ureteral reimplantation (PVUR) has gained popularity due to its minimal invasiveness. However, most of the reported PVUR procedures were based on the Cohen technique. Only few studies reported their experience of PVUR using the Politano-Leadbetter technique (PVUR-PL). Here, we reported our experience of PVUR-PL using a novel technique to facilitate locating the retrovesical ureter during the procedure. Materials and Methods: The medical records of the patients who underwent PVUR-PL between January 2018 and December 2020 in our institution were retrospectively reviewed. The patients were classified into two groups: the modified group that accepted PVUR-PL using our novel technique (using urethral sound to facilitate identifying the retrovesical ureter) and the traditional group that accepted PVUR-PL not using the novel technique. Clinical data were collected retrospectively. Results: There were 22 patients who underwent PVUR-PL, with 13 in the traditional group and nine in the modified group. The mean operating time for unilateral cases in the modified group was significantly shorter than that in the traditional group (154.5 vs. 195.5 min, p < 0.001). For bilateral cases, the mean operating time was also significantly reduced (from 263.0 to 221.3 min, p = 0.022) in the modified group. There were no severe complications in each of the two groups. The peritoneum was perforated in one case from the traditional group, while no peritoneum perforation occurred in the modified group. Conclusion: The use of urethral sound to help to identify the retrovesical ureter during PVUR-PL is a safe and effective technique. This simple but effective technique could shorten the operating time of PVUR-PL and reduce the risk of peritoneum perforation.

7.
Am J Cancer Res ; 11(1): 123-137, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33520364

RESUMEN

Increasing evidence indicates the dysregulations and pivotal roles of lncRNAs in the development and progression of various cancers, including pancreatic cancer. Enhanced glycolytic flux and epithelial-to-mesenchymal transition (EMT) have been considered as important factors in driving the malignance of pancreatic cancer. Here, we sought to evaluate the biological role and involved mechanism of lncRNA CASC9 (CASC9) in pancreatic cancer. Our present study showed that CASC9 was upregulated in various pancreatic cancer cell lines. Loss- and gain-of function of CASC9 demonstrated its critical roles in promoting the glycolysis and EMT phenotypes of pancreatic cancer. Moreover, knockdown of CASC9 inhibited the tumorigenicity and metastasis in vivo. Additionally, our findings showed that hypoxia induced the expression of CASC9 and enhanced the binding of HIF-1α to its promoter. We also demonstrated that the positive feedback loop of CASC9 and the AKT/HIF-1α signaling cascade partially mediated this biological process. Altogether, our results suggest that CASC9 promotes the glycolysis and EMT of pancreatic cancer by a positive feedback loop with AKT/HIF-1α signaling, which is synergistically enhanced by the tumor hypoxic niche. Our study will provide potential therapeutic targets for treating pancreatic cancer.

8.
Front Oncol ; 10: 1411, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32974147

RESUMEN

Background: The spontaneous regression of neuroblastoma (NB) is most prevalent and well-documented in stage 4s NB patients. However, whether autophagy plays roles in the spontaneous regression of NB is unknown. Objective: This study aimed to identify autophagy-related genes (ARGs) and autophagy-related long non-coding RNAs (lncRNAs) differentially expressed in stage 4 and stage 4s NB and to build prognostic risk signatures on the basis of the ARGs and autophagy-related lncRNAs. Methods: One RNA-sequence (RNA-Seq) dataset (TARGET NBL, n = 153) was utilized as discovery cohort, and two microarray datasets (n = 498 and n = 223) were used as validation cohorts. Differentially expressed ARGs were identified by comparing stage 4s and stage 4 NB samples. An ARG signature risk score and an autophagy-related lncRNA signature risk score were constructed. The receiver operating characteristic (ROC) curve analyses were used to evaluate the survival prediction ability of the two signatures. Gene function annotation and Gene Set Enrichment Analysis (GSEA) were performed to clarify the autophagic biological processes enriched in different risk groups. Results: Nine ARGs were integrated into the ARG signature. Patients in the high-risk group of the ARG signature had significantly poorer overall survival (OS) than patients in the low-risk group. The ROC curves analyses revealed that the ARG signature performed very well in predicting OS [5-year area under the curve (AUC) = 0.81]. Seven autophagy-related lncRNAs were integrated into the autophagy-related lncRNA signature. Patients in the high-risk group of the lncRNA signature had significantly poorer OS than patients in the low-risk group. The ROC curve analyses also revealed that the lncRNA signature performed well in predicting OS (5-year AUC = 0.77). Both the ARG signature and lncRNA signature are independent with other clinical risk factors in the multivariate Cox regression survival analyses. GSEAs revealed that autophagy-related biological processes are enriched in low-risk groups. Conclusions: Autophagy-related genes and lncRNAs are differentially expressed between stage 4 and stage 4s NB. The ARG signature and autophagy-related lncRNA signature successfully stratified NB patients into two risk groups. Autophagy-related biological processes are highly enriched in low-risk NB groups.

9.
J Cell Mol Med ; 24(18): 10367-10381, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32683778

RESUMEN

The relationship between metabolism reprogramming and neuroblastoma (NB) is largely unknown. In this study, one RNA-sequence data set (n = 153) was used as discovery cohort and two microarray data sets (n = 498 and n = 223) were used as validation cohorts. Differentially expressed metabolic genes were identified by comparing stage 4s and stage 4 NBs. Twelve metabolic genes were selected by LASSO regression analysis and integrated into the prognostic signature. The metabolic gene signature successfully stratifies NB patients into two risk groups and performs well in predicting survival of NB patients. The prognostic value of the metabolic gene signature is also independent with other clinical risk factors. Nine metabolism-related long non-coding RNAs (lncRNAs) were also identified and integrated into the metabolism-related lncRNA signature. The lncRNA signature also performs well in predicting survival of NB patients. These results suggest that the metabolic signatures have the potential to be used for risk stratification of NB. Gene set enrichment analysis (GSEA) reveals that multiple metabolic processes (including oxidative phosphorylation and tricarboxylic acid cycle, both of which are emerging targets for cancer therapy) are enriched in the high-risk NB group, and no metabolic process is enriched in the low-risk NB group. This result indicates that metabolism reprogramming is associated with the progression of NB and targeting certain metabolic pathways might be a promising therapy for NB.


Asunto(s)
Perfilación de la Expresión Génica , Análisis por Micromatrices , Neuroblastoma/genética , Neuroblastoma/metabolismo , RNA-Seq , 20-Hidroxiesteroide Deshidrogenasas/genética , 20-Hidroxiesteroide Deshidrogenasas/metabolismo , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Anotación de Secuencia Molecular , Mutación/genética , Pronóstico , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Reproducibilidad de los Resultados
10.
Cancer Med ; 9(11): 3800-3815, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32216054

RESUMEN

BACKGROUND: The association between long noncoding RNAs (lncRNAs) and spontaneous regression of neuroblastoma (NB) has rarely been investigated and remains unknown. OBJECTIVE: To identify prognostic lncRNAs involved in the spontaneous regression of NB. METHODS: Differential expression analyses were performed between those samples with an outcome of death in stage 4 NB group and those samples with an outcome of survival in stage 4S NB group in two independent public datasets, respectively. Univariate Cox proportional hazard regression survival analysis was performed in each of the entire cohort to identify those lncRNAs significantly associated with overall survival (OS). Those lncRNAs independently associated with OS were then identified by multivariate Cox survival analysis and used to construct an lncRNA risk score. RESULTS: A total of 20 differentially expressed and survival-related lncRNAs were identified sharing between the two independent cohorts. The expression of each of these 20 lncRNAs was significantly correlated with the expression of NTRK1, which is a well-known factor involved in NB spontaneous regression. Four lncRNAs (LNC00839, FIRRE, LOC283177, and LOC101928100) were identified to be significantly associated with survival independent with each other and a four-lncRNA signature risk score was constructed. Patients with high lncRNA signature risk score had a significantly poorer OS and event-free survival than those with low lncRNA signature risk score. The four-lncRNA signature has a good performance in predicting survival independent with MYCN amplification (nonamplified vs amplified), age status (<18 months vs ≥18 months), risk status (low risk vs high risk), and International Neuroblastoma Staging System (INSS) stage (INSS 1/2/3/4S vs INSS 4). CONCLUSIONS: We identified 20 survival-related lncRNAs that might be associated with the spontaneous regression of NB and developed a four-lncRNA signature risk score. The four-lncRNA signature is an independent prognostic factor for survival of NB patients.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Mutación , Neuroblastoma/genética , Neuroblastoma/patología , ARN Largo no Codificante/genética , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Humanos , Lactante , Masculino , Pronóstico , Tasa de Supervivencia
11.
J Hematol Oncol ; 13(1): 24, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32216806

RESUMEN

BACKGROUND: Aerobic glycolysis is a hallmark of metabolic reprogramming that contributes to tumor progression. However, the mechanisms regulating expression of glycolytic genes in neuroblastoma (NB), the most common extracranial solid tumor in childhood, still remain elusive. METHODS: Crucial transcriptional regulators and their downstream glycolytic genes were identified by integrative analysis of a publicly available expression profiling dataset. In vitro and in vivo assays were undertaken to explore the biological effects and underlying mechanisms of transcriptional regulators in NB cells. Survival analysis was performed by using Kaplan-Meier method and log-rank test. RESULTS: Hepatocyte nuclear factor 4 alpha (HNF4A) and its derived long noncoding RNA (HNF4A-AS1) promoted aerobic glycolysis and NB progression. Gain- and loss-of-function studies indicated that HNF4A and HNF4A-AS1 facilitated the glycolysis process, glucose uptake, lactate production, and ATP levels of NB cells. Mechanistically, transcription factor HNF4A increased the expression of hexokinase 2 (HK2) and solute carrier family 2 member 1 (SLC2A1), while HNF4A-AS1 bound to heterogeneous nuclear ribonucleoprotein U (hnRNPU) to facilitate its interaction with CCCTC-binding factor (CTCF), resulting in transactivation of CTCF and transcriptional alteration of HNF4A and other genes associated with tumor progression. Administration of a small peptide blocking HNF4A-AS1-hnRNPU interaction or lentivirus-mediated short hairpin RNA targeting HNF4A-AS1 significantly suppressed aerobic glycolysis, tumorigenesis, and aggressiveness of NB cells. In clinical NB cases, high expression of HNF4A-AS1, hnRNPU, CTCF, or HNF4A was associated with poor survival of patients. CONCLUSIONS: These findings suggest that therapeutic targeting of HNF4A-AS1/hnRNPU/CTCF axis inhibits aerobic glycolysis and NB progression.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Glucólisis , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Neuroblastoma/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Unión a CCCTC/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo U/genética , Humanos , Neuroblastoma/genética , Neuroblastoma/patología , Mapas de Interacción de Proteínas , ARN Largo no Codificante/genética
12.
Curr Med Sci ; 40(1): 95-103, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32166670

RESUMEN

Helicobacter pylori (H. pylori) was reported to be associated with gastric carcinogenesis. Resistin-like molecule beta (RELMß), a recently described goblet cell-specific protein, was demonstrated to aberrantly express in gastric cancer and correlated with its clinicopathological features. This study aimed to examine the association between H. pylori and RELMß expression in gastric carcinoma and precursor lesions. H. pylori infection and RELMß expression were immunohistochemically evaluated in gastric biopsies from 230 patients. The biopsies consisted of normal gastric mucosa (n=20), mucosa with chronic gastritis (n=41), intestinal metaplasia (n=42), dysplasia (n=31), intestinal-type adenocarcinoma (n=56), and diffuse-type adenocarcinoma (n=40). RELMß expression was measured in gastric biopsies after H. pylori eradication therapy in a subgroup of 32 patients. Cultured gastric cancer cell line SGC-7901 was infected with H. pylori strains, and RELMß expression was detected by reverse transcription PCR, real-time PCR and Western blotting. Higher RELMß immunoreactivity was observed in H. pylori-positive intestinal metaplasia (P=0.003), dysplasia (P=0.032), intestinal-type (P=0.037) and diffuse-type adenocarcinomas (P=0.001) than in H. pylori-negative specimens. Expression rates of RELMß in dysplasia (P=0.005), intestinal-type adenocarcinoma (P<0.001), and diffuse-type adenocarcinoma (P=0.001) were significantly correlated with the grade of H. pylori density. In addition, H. pylori eradication reduced the RELMß intensity in intestinal metaplasia (P=0.001). Infection of gastric cancer SGC-7901 cells with cag pathogenicity island (PAI)-positive H. pylori TN2, but not with its PAI totally deleted mutant (TN2-ΔPAI) for 4-8 h, resulted in enhanced protein and transcript levels of RELMß (P<0.05). In summary, our study suggested that H. pylori infection facilitated the expression of RELMß in gastric garcinoma and precursor lesions.


Asunto(s)
Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/patogenicidad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Gástricas/metabolismo , Regulación hacia Arriba , Adenocarcinoma/metabolismo , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Adulto , Anciano , Amoxicilina/farmacología , Amoxicilina/uso terapéutico , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Metaplasia , Metronidazol/farmacología , Metronidazol/uso terapéutico , Persona de Mediana Edad , Neoplasias Gástricas/microbiología
13.
Theranostics ; 10(4): 1555-1571, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042322

RESUMEN

As a hallmark of metabolic reprogramming, aerobic glycolysis contributes to tumorigenesis and aggressiveness. However, the mechanisms and therapeutic strategies regulating aerobic glycolysis in neuroblastoma (NB), one of leading causes of cancer-related death in childhood, still remain elusive. Methods: Transcriptional regulators and their downstream glycolytic genes were identified by a comprehensive screening of publicly available datasets. Dual-luciferase, chromatin immunoprecipitation, real-time quantitative RT-PCR, western blot, gene over-expression or silencing, co-immunoprecipitation, mass spectrometry, peptide pull-down assay, sucrose gradient sedimentation, seahorse extracellular flux, MTT colorimetric, soft agar, matrigel invasion, and nude mice assays were undertaken to explore the biological effects and underlying mechanisms of transcriptional regulators in NB cells. Survival analysis was performed by using log-rank test and Cox regression assay. Results: Transcription factor myeloid zinc finger 1 (MZF1) was identified as an independent prognostic factor (hazard ratio=2.330, 95% confidence interval=1.021 to 3.317), and facilitated glycolysis process through increasing expression of hexokinase 2 (HK2) and phosphoglycerate kinase 1 (PGK1). Meanwhile, a 21-amino acid peptide encoded by upstream open reading frame of MZF1, termed as MZF1-uPEP, bound to zinc finger domain of Yin Yang 1 (YY1), resulting in repressed transactivation of YY1 and decreased transcription of MZF1 and downstream genes HK2 and PGK1. Administration of a cell-penetrating MZF1-uPEP or lentivirus over-expressing MZF1-uPEP inhibited the aerobic glycolysis, tumorigenesis and aggressiveness of NB cells. In clinical NB cases, low expression of MZF1-uPEP or high expression of MZF1, YY1, HK2, or PGK1 was associated with poor survival of patients. Conclusions: These results indicate that therapeutic targeting of YY1/MZF1 axis by MZF1-uPEP inhibits aerobic glycolysis and NB progression.


Asunto(s)
Terapia Molecular Dirigida/métodos , Neuroblastoma/tratamiento farmacológico , Efecto Warburg en Oncología/efectos de los fármacos , Animales , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Proliferación Celular/genética , Niño , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Hexoquinasa/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Desnudos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Regiones Promotoras Genéticas , Análisis de Supervivencia , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Factor de Transcripción YY1/efectos de los fármacos , Factor de Transcripción YY1/metabolismo
14.
Dis Markers ; 2020: 8736509, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31998420

RESUMEN

BACKGROUNDS: Both pretreatment serum CRP (C-reactive protein) level and ALB (albumin) level have been found to be predictive of survival for multiple malignancies including sarcoma. Since both of the GPS (Glasgow prognostic score) and CAR (C-reactive protein to albumin ratio) are based on the combination of CRP and ALB, we conducted a meta-analysis to evaluate the prognostic role of these two parameters for sarcoma patients. METHODS: A detailed literature search was conducted in MEDLINE, Embase, and Cochrane Library for relevant research publications written in English. Patients' clinical characteristics, outcomes of overall survival (OS), disease-specific survival (DSS), and disease-free survival (DFS) were extracted. Pooled hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were combined to evaluate the prognostic role of GPS or CAR. RESULTS: Twelve articles containing 2695 patients were identified as eligible studies. The results showed that an elevated GPS was significantly correlated with poor OS (HR = 2.42; 95% CI: 1.98-2.94; p < 0.001; fixed-effects model), DSS (HR = 2.28; 95% CI: 1.75-2.97; p < 0.001; fixed-effects model), DSS (HR = 2.28; 95% CI: 1.75-2.97; p < 0.001; fixed-effects model), DSS (HR = 2.28; 95% CI: 1.75-2.97; p < 0.001; fixed-effects model), DSS (HR = 2.28; 95% CI: 1.75-2.97; p < 0.001; fixed-effects model), DSS (HR = 2.28; 95% CI: 1.75-2.97. CONCLUSION: An elevated GPS is predictive of poor survival in patients with sarcomas and is promising to be used as a factor for risk stratification. A higher CAR value is also predictive of poor survival; however, the optimal CAR cut-off value is still to be determined.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteína C-Reactiva/análisis , Sarcoma/sangre , Albúmina Sérica Humana/análisis , Biomarcadores de Tumor/normas , Proteína C-Reactiva/normas , Humanos , Sarcoma/patología , Albúmina Sérica Humana/normas , Análisis de Supervivencia
15.
Mol Ther ; 28(3): 946-962, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31982037

RESUMEN

Recent studies suggest that long noncoding RNAs (lncRNAs) play essential roles in tumor progression. However, the functional roles and underlying mechanisms of lncRNAs in neuroblastoma (NB), the most common malignant solid tumor in pediatric population, still remain elusive. Herein, through integrating analysis of a public RNA sequencing dataset, neuroblastoma highly expressed 1 (NHEG1) was identified as a risk-associated lncRNA, contributing to an unfavorable outcome of NB. Depletion of NHEG1 led to facilitated differentiation and decreased growth and aggressiveness of NB cells. Mechanistically, NHEG1 bound to and stabilized DEAD-box helicase 5 (DDX5) protein through repressing proteasome-mediated degradation, resulting in ß-catenin transactivation that altered target gene expression associated with NB progression. We further determined a lymphoid enhancer binding factor 1 (LEF1)/transcription factor 7-like 2 (TCF7L2)/NHEG1/DDX5/ß-catenin axis with a positive feedback loop and demonstrated that NHEG1 harbored oncogenic properties via its interplay with DDX5. Administration of small interfering RNAs against NHEG1 or DDX5 reduced tumor growth and prolonged survival of nude mice bearing xenografts. High NHEG1 or DDX5 expression was associated with poor survival of NB patients. These results indicate that lncRNA NHEG1 exhibits oncogenic activity that affects NB progression via stabilizing the DDX5 protein, which might serve as a potential therapeutic target for NB.


Asunto(s)
ARN Helicasas DEAD-box/genética , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/genética , ARN Largo no Codificante/genética , beta Catenina/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Biología Computacional , ARN Helicasas DEAD-box/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Modelos Biológicos , Neuroblastoma/metabolismo , Neuroblastoma/mortalidad , Neuroblastoma/patología , Pronóstico , Unión Proteica , Estabilidad del ARN , Factor 1 de Transcripción de Linfocitos T/genética , Activación Transcripcional , beta Catenina/metabolismo
16.
EMBO Mol Med ; 11(12): e10835, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31709724

RESUMEN

Aerobic glycolysis is a hallmark of metabolic reprogramming in tumor progression. However, the mechanisms regulating glycolytic gene expression remain elusive in neuroblastoma (NB), the most common extracranial malignancy in childhood. Herein, we identify that CUT-like homeobox 1 (CUX1) and CUX1-generated circular RNA (circ-CUX1) contribute to aerobic glycolysis and NB progression. Mechanistically, p110 CUX1, a transcription factor generated by proteolytic processing of p200 CUX1, promotes the expression of enolase 1, glucose-6-phosphate isomerase, and phosphoglycerate kinase 1, while circ-CUX1 binds to EWS RNA-binding protein 1 (EWSR1) to facilitate its interaction with MYC-associated zinc finger protein (MAZ), resulting in transactivation of MAZ and transcriptional alteration of CUX1 and other genes associated with tumor progression. Administration of an inhibitory peptide blocking circ-CUX1-EWSR1 interaction or lentivirus mediating circ-CUX1 knockdown suppresses aerobic glycolysis, growth, and aggressiveness of NB cells. In clinical NB cases, CUX1 is an independent prognostic factor for unfavorable outcome, and patients with high circ-CUX1 expression have lower survival probability. These results indicate circ-CUX1/EWSR1/MAZ axis as a therapeutic target for aerobic glycolysis and NB progression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Northern Blotting , Western Blotting , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glucólisis , Células HEK293 , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Técnicas In Vitro , Espectrometría de Masas , Ratones Endogámicos BALB C , Ratones Desnudos , Neuroblastoma , Células PC-3 , Proteína EWS de Unión a ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Factores de Transcripción/genética
17.
Adv Sci (Weinh) ; 6(19): 1900581, 2019 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-31592410

RESUMEN

Proline synthesis plays an important role in the metabolic reprogramming that contributes to tumor progression. However, the mechanisms regulating expression of proline synthetic genes in neuroblastoma (NB) remain elusive. Herein, through integrative screening of a public dataset and amino acid profiling analysis, myeloid zinc finger 1 (MZF1) and MZF1 antisense RNA 1 (MZF1-AS1) are identified as transcriptional regulators of proline synthesis and NB progression. Mechanistically, transcription factor MZF1 promotes the expression of aldehyde dehydrogenase 18 family member A1 and pyrroline-5-carboxylate reductase 1, while proline facilitates the aggressiveness of NB cells. In addition, MZF1-AS1 binds poly(ADP-ribose) polymerase 1 (PARP1) to facilitate its interaction with E2F transcription factor 1 (E2F1), resulting in transactivation of E2F1 and upregulation of MZF1 and other oncogenic genes associated with tumor progression. Administration of a small peptide blocking MZF1-AS1-PARP1 interaction or lentivirus-mediated short hairpin RNA targeting MZF1-AS1 suppresses the proline synthesis, tumorigenesis, and aggressiveness of NB cells. In clinical NB cases, high expression of MZF1-AS1, PARP1, E2F1, or MZF1 is associated with poor survival of patients. These results indicate that therapeutic targeting of MZF1-AS1/PARP1/E2F1 axis inhibits proline synthesis and NB progression.

18.
PLoS One ; 14(7): e0219215, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31260491

RESUMEN

BACKGROUNDS: Serum C-reactive protein (CRP) level has been shown to be a predictor of survival for multiple cancer types. The aim of this study was to evaluate whether pretreatment serum CRP level could serve as a reliable independent prognostic indicator for survival in patients with soft tissue sarcoma (STS). METHODS: A detailed literature search was conducted in Medline, Embase and Cochrane for relevant research publications written in English. Patients' clinical characteristics, outcomes of disease-specific survival (DSS) and disease/recurrence free survival (DFS/RFS) were extracted. Only the results of multivariate survival analysis were recruited in our analysis. Pooled hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were calculated to evaluate the prognostic role of CRP. This study was registered on PROPERO and the registration number is CRD42018104802. RESULTS: Nine articles containing 1655 patients were identified as eligible studies. The random effects model showed that elevated CRP level was significantly correlated with poor DSS (HR = 2.08; 95% CI: 1.33-3.24; p < 0.001). After excluding the heterogeneous study, the fixed effects model showed that elevated CRP level was firmly correlated with poor DSS (HR = 2.36; 95% CI: 1.84-3.03; p < 0.001). The fixed effects model revealed that elevated CRP level was significantly correlated with poor DFS (HR = 1.78; 95% CI: 1.39-2.30; p < 0.001) among studies have more than 100 samples. CONCLUSION: The results of this meta-analysis suggest that elevated pretreatment serum CRP level could serve as an independent risk factor for poor DSS and DFS/RFS in STS patents.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteína C-Reactiva/análisis , Recurrencia Local de Neoplasia/epidemiología , Sarcoma/mortalidad , Supervivencia sin Enfermedad , Humanos , Recurrencia Local de Neoplasia/prevención & control , Pronóstico , Factores de Riesgo , Sarcoma/sangre , Sarcoma/terapia
19.
Oxid Med Cell Longev ; 2019: 8639791, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30931081

RESUMEN

BACKGROUND: As a key step in enhancing cancer cell invasion and metastasis, epithelial-mesenchymal transition (EMT) plays an important role in colorectal cancer progression. EMT is triggered by a variety of signaling pathways, among which the transforming growth factor ß (TGF-ß) signaling pathway has been implicated as a primary inducer. Accumulating evidence demonstrates that MnTE-2-PyP (chemical name: manganese(III) meso-tetrakis-(N-ethylpyridinium-2-yl), a superoxide dismutase (SOD) mimetic, inhibits TGF-ß signaling; however, its ability to inhibit TGF-ß-induced EMT in colorectal cancer has not yet been explored. METHODS: To verify our hypothesis that MnTE-2-PyP attenuates TGF-ß-induced EMT, human colorectal cancer cells were treated with TGF-ß in the presence or absence of MnTE-2-PyP. Cells were analyzed by several techniques including western blotting, real-time quantitative PCR, transwell assay, and wound healing assay. RESULTS: MnTE-2-PyP reverses cell phenotypes induced by TGF-ß in colon cancer cells. MnTE-2-PyP treatment significantly reduced the expression of mesenchymal markers but maintained epithelial marker expression. Mechanistically, MnTE-2-PyP suppressed the phosphorylated Smad2/3 protein levels induced by TGF-ß in SW480 cells, but MnTE-2-PyP failed to suppress TGF-ß-induced Slug and Snail expression in colorectal cells. Furthermore, MnTE-2-PyP effectively suppressed TGF-ß-mediated cell migration and invasion and the expression of matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9) in colorectal cells. CONCLUSION: Taken together, we provide an in-depth mechanism by which MnTE-2-PyP inhibits colorectal cancer progression, supporting an important role for MnTE-2-PyP as an effective and innovative antitumor agent to enhance treatment outcomes in colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/genética , Metaloporfirinas/metabolismo , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
20.
Oxid Med Cell Longev ; 2019: 3643715, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31915505

RESUMEN

BACKGROUND: Metastasis and invasion are the main causes of mortality in gastric cancer. To improve the treatment of gastric cancer, the development of effective and innovative antitumor agents toward invasion and proliferation is needed. Alpha-lipoic acid (ALA), a naturally occurring thiol antioxidant, showed antiproliferative and cytotoxic effects on several cancers. So it is feasible to explore whether ALA can be used to inhibit proliferation and invasion in human gastric cancer. METHODS: The expression of MUC4 in human gastric cancer tissues was assayed by immunohistochemistry. Then, we performed in vitro cell proliferation and invasion analysis to explore the antitumor effect of ALA using AGS, BGC-823, and MKN-28 cells. To further explore the mechanism of ALA-mediated downregulation of MUC4, we cotransfected human gastric cancer cells with STAT3 siRNA and STAT3 overexpression construct. ChIP assays were carried out to find the relationship between MUC4 and STAT3. RESULTS: We found that the MUC4 gene was strongly expressed in human gastric cancer tissues. Meanwhile, ALA reduced proliferation and invasion of human gastric cancer cells by suppressing MUC4 expression. We also found that STAT3 was involved in the inhibition of MUC4 by ALA. Mechanistically, ALA suppressed MUC4 expression by inhibiting STAT3 binding to the MUC4 promoter region. CONCLUSION: ALA inhibits both proliferation and invasion of gastric cancer cells by suppression of STAT3-mediated MUC4 gene expression.


Asunto(s)
Antioxidantes/farmacología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mucina 4/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/metabolismo , Ácido Tióctico/farmacología , Línea Celular Tumoral , Humanos , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA