Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Sci Rep ; 13(1): 13216, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37580432

RESUMEN

Associations between leisure sedentary behavior (especially leisure screen time, LST) and irritable bowel syndrome (IBS) have been reported, but causality is unclear. Here, the two-sample Mendelian randomization was performed to investigate the causal association between LST and IBS. Two recently published genome-wide association studies (GWASs) including a total of 1,190,502 people from Europe were used as our data source. Inverse variance weighting (OR = 1.120, 95% CI 1.029-1.219) and weighted median (OR = 1.112, 95% CI 1.000-1.236) analyses revealed a causal effect between LST and IBS. There was no evidence of pleiotropy in the sensitive analysis (MR-Egger, p = 0.139). After removing potentially confounding single nucleotide polymorphisms (SNPs), similar results were found using inverse variance weighting (OR = 1.131, 95% CI 1.025-1.248) and weighted median (OR = 1.151, 95% CI 1.020-1.299), as well as in the validation analyses using inverse variance weighting (OR = 1.287, 95% CI 0.996-1.662). This study provided support for a possible causal relationship between leisure screen time and IBS.


Asunto(s)
Estudio de Asociación del Genoma Completo , Síndrome del Colon Irritable , Humanos , Síndrome del Colon Irritable/etiología , Síndrome del Colon Irritable/genética , Análisis de la Aleatorización Mendeliana , Tiempo de Pantalla , Causalidad
2.
Heliyon ; 9(1): e12515, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36691542

RESUMEN

Metabolic reprogramming is one of the essential features of tumor that may dramatically contribute to metastasis and collapse. The metabolic profiling is investigated on the patient derived tissue and cancer cell line derived mouse metastasis xenograft. As well-recognized "seeds" for remote metastasis of tumor, role of circulating tumor cells (CTCs) in the study of metabolic reprogramming feature of tumor is yet to be elucidated. More specifically, whether there is difference of metabolic features of liver metastasis in colorectal cancer (CRC) derived from either CTCs or cancer cell line is still unknown. In this study, comprehensive untargeted metabolomics was performed using high performance liquid chromatography-mass spectrometry (HPLC-MS) in liver metastasis tissues from CT26 cells and CTCs derived mouse models. We identified 288 differential metabolites associated with the pathways such as one carbon pool by folate, folate biosynthesis and histidine metabolism through bioinformation analysis. Multiple gene expression was upregulated in the CTCs derived liver metastasis, specifically some specific enzymes. These results indicated that the metabolite phenotype and corresponding gene expression in the CTCs derived liver metastasis tissues was different from the parental CT26 cells, displaying a specific up-regulation of mRNAs involved in the above metabolism-related pathways. The metabolic profile of CTCs was characterized on the liver metastatic process in colorectal cancer. The invasion ability and chemo drug tolerance of the CTCs derived tumor and metastasis was found to be overwhelming higher than cell line derived counterpart. Identification of the differential metabolites will lead to a better understanding of the hallmarks of the cancer progression and metastasis, which may suggest potential attractive target for treating metastatic CRC.

3.
Front Pharmacol ; 13: 816891, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35668932

RESUMEN

Background: The gut microbiota is associated with osteoarthritis (OA) progression. Miya (MY) is a product made from Clostridium butyricum, a member of gut microbiota. This study was conducted to investigate the effects of MY on OA and its underlying mechanisms. Methods: An OA rat model was established, and MY was used to treat the rats for 4 weeks. Knee joint samples from the rats were stained with hematoxylin-eosin, and fecal samples from the OA and OA+MY groups were subjected to 16S rDNA sequencing and metabolomic analysis. The contents of succinate dehydrogenase and muscle glycogen in the tibia muscle were determined, and related genes and proteins were detected using quantitative reverse transcription polymerase chain reaction and western blotting. Results: Hematoxylin and eosin staining showed that treatment with MY alleviated the symptoms of OA. According to the sequencing results, MY significantly increased the Chao1, Shannon, and Pielou evenness values compared to those in the untreated group. At the genus level, the abundances of Prevotella, Ruminococcus, Desulfovibrio, Shigella, Helicobacter, and Streptococcus were higher in the OA group, whereas Lactobacillus, Oscillospira, Clostridium, and Coprococcus were enriched after MY treatment. Metabolomic analysis revealed 395 differentially expressed metabolites. Additionally, MY treatment significantly increased the succinate dehydrogenase and muscle glycogen contents in the muscle caused by OA (p > 0.05). Finally, AMPK, Tfam, Myod, Ldh, Chrna1, Chrnd, Rapsyn, and Agrin were significantly downregulated in the muscles of OA mice, whereas Lcad, Mcad, and IL-1ß were upregulated; MY significantly reversed these trends induced by OA. Conclusions: MY may promote the repair of joint damage and protect against OA via the gut-muscle-joint axis.

4.
Front Immunol ; 13: 828243, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711417

RESUMEN

Programmed cell death (PCD) plays an important role in the onset and progression of various cancers. The molecular events surrounding the occurrence of abnormally expressed long noncoding RNAs (lncRNAs) leading to colon cancer (CC) have become a focus. We comprehensively evaluated the roles of PCD-related lncRNAs in the clinical management of CC and their immune responses. Therefore, we screened 41 prognostic PCD-related lncRNAs in The Cancer Genome Atlas database using co-expression analysis and assigned patients to groups according to the results of cluster analysis. The immune response and functions of cluster 2 were substantially suppressed, which might explain the poor prognosis in this group. A prognostic model comprising eight PCD-related lncRNAs was developed, and its effectiveness was verified using an external database. High-and low-risk groups had different epigenetic modifications and changes in immune cell infiltration. Patients in the high-risk group were resistant to immunotherapy and various chemotherapeutic drugs. Studies in vitro and in vivo further confirmed a carcinogenic role of the lncRNA U62317.4. Our findings of the prognostic value of PCD-related lncRNAs revealed their important roles in immune response disorders, thus providing valuable insights into the clinical management and molecular mechanisms of CC.


Asunto(s)
Neoplasias del Colon , ARN Largo no Codificante , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/terapia , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Pronóstico , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
Ann Transl Med ; 10(2): 94, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35282090

RESUMEN

Background: A novel colorectal cancer center (CCC) was developed in the Shanghai Tenth People's hospital of Tongji University during the COVID-19 epidemic. In this study, we aimed to evaluate the CCC model in terms of three aspects. Methods: This retrospective study used data from the Shanghai Tenth People's hospital patient databases. The research hypothesis was that the CCC reduces preoperative waiting time (PWT), length of hospital stay (LOS), and costs of hospitalization, without reducing the quality of surgery. Thus, we compared the time, cost, and quality between March 1 to December 31, 2019, and March 1 to December 31, 2020. Descriptive and inferential analyses of patient demographic characteristics, time, postoperative outcomes, and inpatient costs were conducted. Results: A total of 965 hospitalizations for colorectal cancer (CRC) were identified-415 in 2019 and 550 in 2020. In the CCC, PWT declined by 26.2 hours (P<0.01). Patients in the CCC express group only needed to wait for 24.5 hours before undergoing surgery, with a shorter LOS than the normal group (P<0.01). None of the patients had any symptoms of COVID-19 or were high-risk COVID-19 contacts, and the incidence of immediate postoperative complications was low. The mean total inpatient cost (TIC) for all patients with CRC was 78,309.824 Chinese Yuan in 2020, which was slightly lower than that in 2019. Conclusions: This study found that the centralized management model for CRC care could help patients save the PWT, LOS and costs of hospitalization during the COVID-19 epidemic.

6.
Cell Death Discov ; 7(1): 272, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34601499

RESUMEN

Colorectal cancer (CRC) is a common tumor that harms human health with a high recurrence rate. It has been reported that the expression of microRNA-539 (miR-539) is low in several types of cancer, including CRC. Tumor necrosis factor (TNF)-α-induced protein 8 (TNFAIP8/TIPE) is highly expressed in CRC and promotes the proliferation, migration and angiogenesis of CRC. However, the relationship between miR-539 and TIPE and the mechanisms by which they regulate the proliferation of CRC remain to be explored. We aimed to investigate the functions and mechanisms of miR-539 in CRC proliferation. Functionally, miR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. Our data reveal the novel role of miR-539 in regulating ferroptosis in CRC via activation of the SAPK/JNK axis, providing new insight into the mechanism of abnormal proliferation in CRC and a novel potential therapeutic target for advanced CRC.

7.
Front Cell Dev Biol ; 9: 684322, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422808

RESUMEN

The dysregulated expression of glycolysis-related genes (GRGs) is closely related to the occurrence of diverse tumors and regarded as a novel target of tumor therapy. However, the role of GRGs in colon cancer is unclear. We obtained 226 differential GRGs (DE-GRGs) from The Cancer Genome Atlas (TCGA) database. Cox regression analysis was used to construct a DE-GRG prognostic model, including P4HA1, PMM2, PGM2, PPARGC1A, PPP2CB, STC2, ENO3, and CHPF2. The model could accurately predict the overall survival rate of TCGA and GSE17536 patient cohorts. The risk score of the model was closely related to a variety of clinical traits and was an independent risk factor for prognosis. Enrichment analysis revealed the activation of a variety of glycolysis metabolism and immune-related signaling pathways in the high-risk group. High-risk patients displayed low expression of CD4+ memory resting T cells and resting dendritic cells and high expression of macrophages M0 compared with the expression levels in the low-risk patients. Furthermore, patients in the high-risk group had a higher tumor mutation load and tumor stem cell index and were less sensitive to a variety of chemotherapeutic drugs. Quantitative reverse transcription polymerase chain reaction and immunohistochemistry analyses validated the expression of eight GRGs in 43 paired clinical samples. This is the first multi-omics study on the GRGs of colon cancer. The establishment of the risk model may benefit the prognosis and drug treatment of patients.

8.
Surg Endosc ; 35(1): 476-485, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32989539

RESUMEN

BACKGROUND: In patients with ultralow rectal cancer, surgical resection of the tumor without impairing sphincter function remains a technical challenge. The purpose of this study was to describe a new technique of transanal natural orifice specimen extraction (NOSE) surgery using our independently developed devices, aiming to achieve precise cancer resection and preserve sphincter function in patients with ultralow rectal cancer. METHODS: Precision functional sphincter-preserving surgery (PPS) was performed on nineteen patients with ultralow rectal cancer between June 2019 and April 2020. With the help of our independently developed devices, surgeons directly and accurately removed the lower edge of the tumor and retained healthy rectal tissue on the nontumorous side. Hand-sewn anastomosis with a mattress suture was used to achieve sturdy anastomosis. Preoperative baseline characteristics, operative details, 90-day postoperative complications, costs, and anal function score at 6 months after surgery were documented. RESULTS: Nineteen ultralow rectal cancer patients with a median distance to the dentate line of 2.0 cm successfully underwent PPS without serious postoperative complications. Six out of nineteen patients (31.6%) received a prophylactic stoma. The average cost was 62164.1 yuan. At 6 months after surgery, the average Wexner anal function score and the average Vaizey score were both 3 points. CONCLUSIONS: PPS can be employed to precisely resect rectal tumors and preserve sphincter function in ultralow rectal cancer patients. The use of our devices enhanced surgical efficiency, reduced the need for prophylactic stoma, reduced surgery-related costs, and prevented abdominal surgical incisions.


Asunto(s)
Cirugía Endoscópica por Orificios Naturales/métodos , Neoplasias del Recto/cirugía , Anciano , Canal Anal/cirugía , Anastomosis Quirúrgica/métodos , Humanos , Masculino , Persona de Mediana Edad , Cirugía Endoscópica por Orificios Naturales/efectos adversos , Cirugía Endoscópica por Orificios Naturales/instrumentación , Tratamientos Conservadores del Órgano , Complicaciones Posoperatorias/etiología , Recto/cirugía , Estomas Quirúrgicos
10.
ACS Appl Mater Interfaces ; 12(4): 4193-4203, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31935069

RESUMEN

Isolation of circulating tumor cells (CTCs) in peripheral blood from cancer patients bears critical importance for evaluation of therapeutic efficacy. The current CTC isolation strategies are majorly relying on either protein biomarkers or dimensional features of CTCs. In this study, we present a new methodology for CTC detection and isolation based on the surface charge of cancer cells, a bioelectrical manifestation of the "Warburg effect." Negative surface charge is a direct consequence of glycolysis of cancer cells, which can be utilized as an effective biophysical marker for CTC detection and isolation. Upon cancer cells-nanoparticle interaction via optimum incubation, serum protein-coated electrically charged nanoparticles can trap different cancer cells independent of their epithelial protein expression. In fetal bovine serum , the poly(ethyleneimine)-functionalized Fe3O4 nanoparticles, surface-decorated with protein corona, are able to efficiently capture CTCs from blood samples of colorectal cancer patients.  2-8 CTCs has been isolated from 1 mL of blood and identified by immunostaining fluorescence in situ hybridization and immunofluorescence staining in all 25 colorectal cancer patients at varied stages, while only 0-1 CTC was detected from blood samples of 10 healthy donors. Diverse CTC subpopulations of heteroploids and biomarker expression can also be detected in this strategy. The label-free, charge-based CTC method shows promise in cancer diagnosis and prognosis paving a new path for liquid biopsy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Separación Celular , Nanopartículas de Magnetita/química , Células Neoplásicas Circulantes , Corona de Proteínas/química , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología
11.
Front Oncol ; 10: 623048, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33718119

RESUMEN

Tumor necrosis factor-induced protein-8 (TIPE) is highly expressed in colorectal cancer (CRC). Decoy receptor 3 (DcR3) is a soluble secreted protein that can antagonize Fas ligand (FasL)-induced apoptosis and promote tumorigenesis. It remains unclear whether TIPE can regulate DcR3 expression. In this study, we examined this question by analyzing the relationship between these factors in CRC. Bioinformatics and tissue microarrays were used to determine the expression of TIPE and DcR3 and their correlation in CRC. The expression of TIPE and DcR3 in colon cancer cells was detected. Plasma samples were collected from CRC patients, and DcR3 secretion was measured. Then, dual-luciferase reporter gene analysis was performed to assess the interaction between TIPE and DcR3. We exogenously altered TIPE expression and analyzed its function and influence on DcR3 secretion. Lipopolysaccharide (LPS) was used to stimulate TIPE-overexpressing HCT116 cells, and alterations in signaling pathways were detected. Additionally, inhibitors were used to confirm molecular mechanisms. We found that TIPE and DcR3 were highly expressed in CRC patients and that their expression levels were positively correlated. DcR3 was highly expressed in the plasma of cancer patients. We confirmed that TIPE and DcR3 were highly expressed in HCT116 cells. TIPE overexpression enhanced the transcriptional activity of the DcR3 promoter. TIPE activated the PI3K/AKT signaling pathway to regulate the expression of DcR3, thereby promoting cell proliferation and migration and inhibiting apoptosis. In summary, TIPE and DcR3 are highly expressed in CRC, and both proteins are associated with poor prognosis. TIPE regulates DcR3 expression by activating the PI3K/AKT signaling pathway in CRC, thus promoting cell proliferation and migration and inhibiting apoptosis. These findings may have clinical significance and promise for applications in the treatment or prognostication of CRC.

12.
Invest New Drugs ; 38(2): 221-228, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-30887254

RESUMEN

Colorectal cancer (CRC) is the most frequently encountered malignancy associated with the rectum or colon, and accumulating evidences have implicated intestinal dysbacteriosis (IDB, disruption of gut microbiome) and exosomes in the pathology of CRC. We aimed to investigate the effect of IDB on exosome secretion in a CRC xenograft mouse model. An IDB mouse model was established and was inoculated with the CRC cell line SW480 as a xenograft tumor. Tumor growth was monitored for 15 days in sham and IDB mice, after which blood was collected to assess serum exosome secretion. A novel exosome secretion inhibitor, neticonazole, was administered to IDB mice bearing CRC xenograft tumors, followed by monitoring of tumor growth and mouse survival. Western blot analysis was performed in xenograft tumors to investigate the underlying molecular mechanism. IDB promoted CRC xenograft tumor growth and exosome secretion, which could be inhibited by the exosome secretion inhibitor neticonazole. Moreover, neticonazole treatment significantly improved the survival of IDB mice with CRC xenograft tumors, likely through increasing apoptosis of CRC xenograft tumor cells. The exosome secretion inhibitor neticonazole may serve as a promising therapeutic candidate against CRC by suppressing IDB-induced CRC tumorigenesis.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Disbiosis/tratamiento farmacológico , Exosomas/efectos de los fármacos , Imidazoles/uso terapéutico , Intestinos/microbiología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Disbiosis/complicaciones , Humanos , Imidazoles/farmacología , Masculino , Ratones Endogámicos C57BL
13.
Nutrition ; 70: 110498, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31655470

RESUMEN

OBJECTIVE: Although there is international consensus regarding the importance of cachexia, no tools exist, to our knowledge, for cachexia screening among patients with cancer. The aim of this study was to evaluate whether patients with cancer and cachexia could be identified using the four most commonly used nutritional screening tools: the Malnutrition Universal Screening Tool (MUST), the Nutritional Risk Screening (NRS)-2002, the Malnutrition Screening Tool (MST), and the Short Nutritional Assessment Questionnaire (SNAQ). METHODS: Clinical data were prospectively collected for patients who underwent elective radical gastrectomy for gastric cancer in two large centers between August 2014 and February 2018. Patients were also screened using the MUST, NRS-2002, MST, and SNAQ tools. The screening results were subsequently compared with the international consensus diagnostic criteria for cancer cachexia. RESULTS: A total of 1001 patients were evaluated, including 363 patients (36.3%) with cancer cachexia. Among the patients "at nutritional risk" based on each tool, the proportions of cachexia were 87.3% for the MUST tool, 84.3% for the MST tool, 76.6% for the NRS-2002 tool, and 54.3% for the SNAQ tool. The MST tool provided the largest area under the curve for identifying cancer cachexia (0.914; P < 0.001). CONCLUSION: Among the tools examined, the MST had the greatest ability to detect cancer cachexia among patients with gastric cancer.


Asunto(s)
Caquexia/diagnóstico , Desnutrición/diagnóstico , Tamizaje Masivo/métodos , Evaluación Nutricional , Complicaciones Posoperatorias/diagnóstico , Neoplasias Gástricas/complicaciones , Anciano , Antropometría , Índice de Masa Corporal , Caquexia/etiología , Femenino , Gastrectomía/efectos adversos , Humanos , Masculino , Desnutrición/etiología , Persona de Mediana Edad , Estado Nutricional , Complicaciones Posoperatorias/etiología , Estudios Prospectivos , Reproducibilidad de los Resultados , Medición de Riesgo , Neoplasias Gástricas/fisiopatología , Neoplasias Gástricas/cirugía
14.
Oncol Rep ; 42(5): 2117-2129, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31545467

RESUMEN

It has been reported that kruppel­like factor 17 (KLF17) acts as a tumour suppressor in several tissues and cancer cells, however, the molecular roles, the underlying mechanisms and clinical significance of KLF17 in colorectal cancer (CRC) have not been completely elucidated. In the present study, KLF17 protein expression was detected in 140 primary CRCs and paired adjacent non­tumour tissues using immunohistochemistry with tissue microarrays. The KLF17 mRNA expression was determined in 4 CRC cell lines and 20 pairs of the aforementioned tissues using reverse transcription quantitative polymerase chain reaction. The correlation between KLF17 expression and clinicopathologic characteristics was determined. Next, the functions of KLF17 in CRC were examined by cell proliferation, colony formation, adhesion, invasion and mouse xenograft assays. Methylation­specific PCR and bisulfite sequencing PCR were also carried out to investigate the promoter methylation status of KLF17 in CRC cells and tissues and explore the effects of lentiviral­mediated RNAi of UHRF1 on the methylation and expression of KLF17. The results revealed that KLF17 expression was abnormally decreased in CRC and associated with lymph node metastasis and unfavorable overall survival. Moreover, ectopic KLF17 expression suppressed CRC cell growth and invasion in vitro and in vivo. In addition, the downregulation of KLF17 was associated with the hypermethylation of the CpG nucleotides on the KLF17 promoter. The knockdown of the epigenetic regulator UHRF1 reduced the methylation level of the KLF17 promoter and inhibited CRC cell adhesion, invasion and epithelial­mesenchymal transition by upregulating KLF17. The present findings indicated that KLF17 may act as a tumour suppressor gene in CRC and a potential independent prognostic biomarker in CRC patients. UHRF1 can suppress KLF17 expression through the hypermethylation of its promoter in CRC. These results offer insights into the KLF17 expression regulation in CRC and suggest an inhibitory effect of KLF17 on tumourigenesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células CACO-2 , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Análisis de Supervivencia , Análisis de Matrices Tisulares
15.
Surg Endosc ; 33(10): 3384-3395, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30604263

RESUMEN

BACKGROUND: The purpose of the study was to investigate the proliferation and migration capability of human gastrointestinal stromal tumor line GIST-T1 after exposure to different pressures and times of CO2 pneumoperitoneum. METHODS: We established simulated CO2 pneumoperitoneum environment in vitro and divided the human GIST cell GIST-T1 into open control group, 8 mmHg CO2 pneumoperitoneum treatment group and 15 mmHg CO2 pneumoperitoneum treatment group. Each group was divided into two subgroups respectively cultured for 1 h and 3 h. pH value of cell culture, cell growth curve, and cell cycle distribution of each group was measured. By application of scratch healing tests and Transwell chamber experiments, mobility ratio and number of cells through 8 µm membranes were measured to assess the migration ability of cells in each group after intervention. RESULTS: Cell culture pH value of each subgroup in CO2 group decreased significantly after exposed in CO2 pneumoperitoneum (P < 0.01). The proliferation of GIST-T1 cells in 15 mmHg CO2 group was significantly inhibited early (1-2 days) (P < 0.05) and the proliferation of GIST-T1 cells in 8 mmHg CO2 1 h subgroup and 15 mmHg CO2 1 h subgroup was increased significantly late (4-6 days) (P < 0.05) after the interventions of CO2 pneumoperitoneum. The percentage of cells in G0-G1 phase increased, the percentage of S phase cells decreased (P < 0.01) in 1-h subgroup and 3-h subgroup of 15 mmHg CO2 group 24 h after exposure to CO2. The percentage of cells in S phase increased in 1-h subgroup of 8 mmHg CO2 group and decreased in 3-h subgroup of 15 mmHg CO2 group 72 h after exposure to CO2. In the Transwell chamber experiment, the cell number through 8-µm membrane increased significantly (P < 0.01) in 3-h subgroup of CO2 group compared to that in 3-h subgroup of control group. CONCLUSIONS: The routine pressure and duration of CO2 pneumoperitoneum used in clinic did not promote the proliferation of gastrointestinal stromal tumors, but had a potential risk of increasing postoperative recurrence and distant metastasis.


Asunto(s)
Apoptosis , Movimiento Celular , Proliferación Celular , Tumores del Estroma Gastrointestinal/patología , Neumoperitoneo Artificial , Dióxido de Carbono , Línea Celular Tumoral , Humanos
16.
Cell Physiol Biochem ; 48(6): 2337-2349, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30114690

RESUMEN

BACKGROUND/AIMS: Long noncoding RNAs (lncRNAs) constitute a large proportion of noncoding transcripts that have recently emerged as a new class of important regulators in cancers. LncRNA BCYRN1, also known as BC200, has a potential function in tumorigenesis. However, the clinical significance of BCYRN1 and its effect on colorectal cancer (CRC) progression remains unclear. METHODS: Quantitative reverse transcriptase PCR (qRT-PCR) was performed to investigate the expression of BCYRN1 in CRC tissues and cell lines. The biological function of BCYRN1 was also investigated through knockdown and overexpression of BCYRN1 in vitro. Microarray bioinformatics analysis was performed to analyze the putative targets of BCYRN1. RESULTS: The results showed that BCYRN1 expression was significantly upregulated in 96 CRC tumor tissues compared with para-carcinoma control tissues. Additionally, BCYRN1 overexpression was associated with larger tumor size and advanced pathological stages in CRC patients. In vitro BCYRN1 knockdown significantly inhibited the proliferation and apoptosis of CRC cells. Furthermore, NPR3 was identified to be a target of BCYRN1 and was downregulated by BCYRN1 knockdown. CONCLUSION: Together, we provide the first evidence that BCYRN1 plays an oncogenic role in CRC cells. BCYRN1 may be a promising prognostic biomarker and a potential therapeutic target for CRC.


Asunto(s)
Neoplasias Colorrectales/patología , ARN Largo no Codificante/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Receptores del Factor Natriurético Atrial/antagonistas & inhibidores , Receptores del Factor Natriurético Atrial/genética , Transcriptoma , Regulación hacia Arriba
17.
Cancer Cell ; 34(1): 103-118.e9, 2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-30008322

RESUMEN

YAP, a key effector of Hippo pathway, is activated by its translocation from cytoplasm to nucleus to regulate gene expression and promote tumorigenesis. Although the mechanism by which YAP is suppressed in cytoplasm has been well-studied, how the activated YAP is sequestered in the nucleus remains unknown. Here, we demonstrate that YAP is a nucleocytoplasmic shuttling protein and its nuclear export is controlled by SET1A-mediated mono-methylation of YAP at K342, which disrupts the binding of YAP to CRM1. YAP mimetic methylation knockin mice are more susceptible to colorectal tumorigenesis. Clinically, YAP K342 methylation is reversely correlated with cancer survival. Collectively, our study identifies SET1A-mediated mono-methylation at K342 as an essential regulatory mechanism for regulating YAP activity and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/enzimología , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias Pulmonares/enzimología , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Células A549 , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Núcleo Celular/genética , Núcleo Celular/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células HEK293 , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisina , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , Pronóstico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Factores de Transcripción , Carga Tumoral , Proteínas Señalizadoras YAP
18.
Cancer Med ; 7(4): 1253-1263, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29516678

RESUMEN

Long noncoding RNAs (lncRNAs) have emerged as regulators in a variety of biological processes, including carcinogenesis in human cancer. UCA1 has been reported to be upregulated in gastric cancer (GC); however, the underlying functional roles of UCA1 in GC have not been established. In the current study, we showed that UCA1 is significantly higher in GC tissues and cells compared with adjacent normal tissues and a gastric epithelium cell line, respectively. Higher UCA1 expression was associated with lymph node metastasis, TNM stage, and poor overall survival (OS) in GC patients. In vitro functional studies confirmed that UCA1 promotes cell proliferation, colony formation ability, and cell invasion in GC cells. We demonstrated that knockdown of UCA1 inhibits tumor growth in vivo. The double luciferase reporter, RNA-binding protein immunoprecipitation assay, and RNA pull down assay demonstrated that miR-590-3p serves as a target for UCA1. UCA1 promoted cell proliferation and invasion by negatively regulating miR-590-3p expression. Moreover, we demonstrated that CREB1 is a downstream target of miR-590-3p and UCA1 activates CREB1 expression by sponging to miR-590-3p. Thus, these results showed that UCA1 functions as an oncogene in GC and may be a target for treatment of GC.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Interferencia de ARN , ARN Largo no Codificante/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
19.
Onco Targets Ther ; 9: 4473-81, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27524904

RESUMEN

BACKGROUND: TNFAIP8, also known as TIPE, is a suppressor of apoptosis. High expression of both TIPE mRNA and protein has been detected in various cancer cell lines and clinical specimens compared to healthy tissues. Many reports have shown that there is a strong correlation between TIPE overexpression and cancer progression and poor prognosis in human solid cancers. METHODS: To illustrate the functional and clinical significance of TIPE in gastric cancer, we used reverse transcription polymerase chain reaction, quantitative real-time polymerase chain reaction, and immunohistochemistry to measure TIPE expression in clinical gastric specimens. Then, TIPE expression was knocked down by using shRNA and anti-DR5ScFv, to examine different expressions of TIPE in BGC823 cell lines, while cell proliferation and apoptosis were induced. RESULTS: We found that there was a strong correlation between TIPE expression and TNM stage (P=0.044), tumor depth (P=0.016), lymph node metastasis (P=0.026), and distant metastasis (P=0.045). No significant correlation was found between TIPE expression with the patients' age (P=0.062) or sex (P=0.459). Anti-DR5ScFv induced TIPE depletion both in vitro and in vivo and resulted in apoptosis and suppression of proliferation. CONCLUSION: Our results suggested that TIPE expression was associated with gastric cancer progression, and most importantly, suppressing TIPE expression might be an effective therapeutic strategy.

20.
Oncol Lett ; 11(3): 1835-1840, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26998086

RESUMEN

Tumor necrosis factor (TNF)-α-induced protein 8 (TIPE) is a recently identified protein that is considered to be associated with various malignancies, including esophageal, breast and pancreatic cancer; however, the importance of TIPE in gastric cancer (GC) remains unknown. Decoy receptor 3 (DcR3) is a member of the tumor necrosis factor receptor superfamily that is expressed in digestive system neoplasms. The expression of DcR3 is regulated by the mitogen-activated protein kinase (MAPK)/MAPK kinase/extracellular signal-regulated kinase (ERK) signaling pathway. Reverse transcription-polymerase chain reaction was performed to detect the expression of TIPE, ERK and DcR3 in the pathological and tumor-adjacent normal gastric tissues of 30 patients that demonstrated stage III gastric adenocarcinoma. The expression and distribution of the TIPE protein was examined using immunohistochemistry, and the clinical significance and expression levels of DcR3 and ERK1/2 were evaluated. The expression of TIPE, ERK1/2 and DcR3 in the tumor tissues of GC was significantly increased compared with paracarcinoma tissues (P<0.05). In addition, TIPE expression positively correlated with DcR3 and ERK1 levels (r=0.538 and r=0.462, respectively; P<0.05). There was no statistical difference between tumor tissues from patients with varying age, gender, differentiation or lymph node metastasis (P>0.05). TIPE may be vital in the progression of GC. TIPE may be associated with the expression of DcR3 and ERK1/2, which may be involved in the cell apoptosis of GC. The present study elucidates the potential function of TIPE as a novel marker and therapeutic target for GC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...