Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Opt Express ; 32(11): 19388-19396, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38859074

ABSTRACT

A novel fiber Bragg grating (FBG) sensing system, based on an optically injected distributed feedback laser diode (DFB-LD) with an optoelectronic oscillating (OEO) loop, is proposed and experimentally demonstrated for temperature measurements with high and tunable sensitivity. The FBG sensor device works as an edge filter to adjust the optical power of the injected beam in response to temperature variations. The optically injected DFB-LD works at Period-one (P1) oscillating state, and the central wavelength of the oscillating mode of the DFB-LD can be tuned by the variable power of the injected beam. Furthermore, an OEO loop is implemented to improve the signal quality of the generated P1 microwave signal. Hence, the sensing parameter of temperature is converted to the frequency variation of the generated P1 microwave signal in the proposed sensing system. In the proof-of-concept experiment, a series of P1 microwave signals are generated while different temperatures are applied to the FBG sensor. The sensitivity of the proposed FBG sensing system for temperature measurements can be tuned from 0.44322 GHz/°C to 1.25952 GHz/°C. The stability and repeatability experiments are also performed, demonstrating the high measurement accuracy (0.0629°C) and low error of the system. The proposed FBG-based sensing and interrogation system exhibits high sensitivity, large tunability, good linearity, and flexible sensing generality.

2.
BMC Med ; 22(1): 14, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38195495

ABSTRACT

BACKGROUND: In the post-pandemic era, a wide range of COVID-19 sequelae is of growing health concern. However, the risks of digestive diseases in long COVID have not been comprehensively understood. To investigate the long-term risk of digestive diseases among COVID patients. METHODS: In this large-scale retrospective cohort study with up to 2.6 years follow-up (median follow-up: 0.7 years), the COVID-19 group (n = 112,311), the contemporary comparison group (n = 359,671) and the historical comparison group (n = 370,979) predated the COVID-19 outbreak were built using UK Biobank database. Each digestive outcome was defined as the diagnosis 30 days or more after the onset of COVID-19 infection or the index date. Hazard ratios (HRs) and corresponding 95% confidence intervals (CI) were computed utilizing the Cox regression models after inverse probability weighting. RESULTS: Compared with the contemporary comparison group, patients with previous COVID-19 infection had higher risks of digestive diseases, including gastrointestinal (GI) dysfunction (HR 1.38 (95% CI 1.26 to 1.51)); peptic ulcer disease (HR 1.23 (1.00 to 1.52)); gastroesophageal reflux disease (GERD) (HR 1.41 (1.30 to 1.53)); gallbladder disease (HR 1.21 (1.06 to 1.38)); severe liver disease (HR 1.35 (1.03 to 1.76)); non-alcoholic liver disease (HR 1.27 (1.09 to 1.47)); and pancreatic disease (HR 1.36 (1.11 to 1.66)). The risks of GERD were increased stepwise with the severity of the acute phase of COVID-19 infection. Even after 1-year follow-up, GERD (HR 1.64 (1.30 to 2.07)) and GI dysfunction (HR 1.35 (1.04 to 1.75)) continued to pose risks to COVID-19 patients. Compared to those with one SARS-CoV-2 infection, reinfected patients were at a higher risk of pancreatic diseases (HR 2.57 (1.23 to 5.38)). The results were consistent when the historical cohort was used as the comparison group. CONCLUSIONS: Our study provides insights into the association between COVID-19 and the long-term risk of digestive system disorders. COVID-19 patients are at a higher risk of developing digestive diseases. The risks exhibited a stepwise escalation with the severity of COVID-19, were noted in cases of reinfection, and persisted even after 1-year follow-up. This highlights the need to understand the varying risks of digestive outcomes in COVID-19 patients over time, particularly those who experienced reinfection, and develop appropriate follow-up strategies.


Subject(s)
COVID-19 , Digestive System Diseases , Gastroesophageal Reflux , Liver Diseases , Humans , Post-Acute COVID-19 Syndrome , COVID-19/epidemiology , Cohort Studies , Reinfection , Retrospective Studies , SARS-CoV-2 , Digestive System Diseases/epidemiology
3.
Sci Rep ; 13(1): 18660, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37907757

ABSTRACT

In this paper, a structured light vision measurement method using a scanning laser line and a positioning laser line is proposed. The novel method enables the scanning laser plane to slide along a slide rail while maintaining intersection with the positioning laser plane, eliminating the need to determine the scanning direction and moving step. During the measurement process, the laser plane equations need to be recalibrated for each new position, so a real-time calibration method is given. Initially, the geometric barycenter method is employed to detect the subpixel coordinates of the light stripe intersection point. Subsequently, these coordinates are projected into the camera coordinate system using the initial equations of the positioning laser plane. Finally, leveraging the normal information of the initial equation of the scanning laser plane and the three-dimensional coordinates of the light stripe intersection point, the real-time calibration of the scanning laser plane equations can be accomplished. The proposed method enables the three-dimensional reconstruction of objects, and its accuracy is verified through measurements on gauge blocks. Experimental results demonstrate that this method achieves precise and stable three-dimensional reconstruction of object surface shape.

4.
Commun Biol ; 6(1): 1026, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37816820

ABSTRACT

Various miRNAs have been shown to participate in the tumor progression and development of colorectal cancer (CRC). However, the role of miR-3913-5p in CRC are yet to be clearly defined. In the present study, we determine that miR-3913-5p is downregulated in CRC cell lines and CRC tissues. Exogenous miR-3913-5p expression weakens the CRC cells growth, migration and invasion. Mechanistically, miR-3913-5p directly targets the 3'UTR of CREB5. Overexpression of CREB5 reverses the suppression of CRC cells proliferation, migration and invasion induced by miR-3913-5p. Furthermore, ATF2 negatively regulates the transcription of miR-3913-5p by binding to its promoter. CREB5 can cooperate with ATF2. CREB5 is required for ATF2 in regulating miR-3913-5p. Finally, inverse correlations can be found between the expressions of miR-3913-5p and CREB5 or ATF2 in CRC tissues. Thus, a plausible mechanism of ATF2/miR-3913-5p/CREB5 axis regulating CRC progression is elucidated. Our findings suggest that miR-3913-5p functions as a tumor suppressor in CRC. ATF2/miR-3913-5p/CREB5 axis might be a potential therapeutic target against CRC progression.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , Colorectal Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line , Cell Proliferation/genetics , Activating Transcription Factor 2/genetics , Cyclic AMP Response Element-Binding Protein A
5.
World J Gastrointest Oncol ; 15(8): 1384-1399, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37663941

ABSTRACT

BACKGROUND: Altered miR-188-3p expression has been observed in various human cancers. AIM: To investigate the miR-188-3p expression, its roles, and underlying molecular events in gastric cancer. METHODS: Fifty gastric cancer and paired normal tissues were collected to analyze miR-188-3p and CBL expression. Normal and gastric cancer cells were used to manipulate miR-188-3p and CBL expression through different assays. The relationship between miR-188-3p and CBL was predicted bioinformatically and confirmed using a luciferase gene reporter assay. A Kaplan-Meier analysis was used to associate miR-188-3p or CBL expression with patient survival. A nude mouse tumor cell xenograft assay was used to confirm the in vitro data. RESULTS: MiR-188-3p was found to be lower in the plasma of gastric cancer patients, tissues, and cell lines compared to their healthy counterparts. It was associated with overall survival of gastric cancer patients (P < 0.001), tumor differentiation (P < 0.001), lymph node metastasis (P = 0.033), tumor node metastasis stage (I/II vs III/IV, P = 0.024), and American Joint Committee on Cancer stage (I/II vs III/IV, P = 0.03). Transfection with miR-188-3p mimics reduced tumor cell growth and invasion while inducing apoptosis and autophagy. CBL was identified as a direct target of miR-188-3p, with its expression antagonizing the effects of miR-188-3p on gastric cancer (GC) cell proliferation by inducing tumor cell apoptosis and autophagy through the inactivation of the Akt/mTOR signaling pathway. The in vivo data confirmed antitumor activity via CBL downregulation in gastric cancer. CONCLUSION: The current data provides ex vivo, in vitro, and in vivo evidence that miR-188-3p acts as a tumor suppressor gene or possesses antitumor activity in GC.

7.
Oncogene ; 41(43): 4823-4838, 2022 10.
Article in English | MEDLINE | ID: mdl-36153370

ABSTRACT

Although the abnormal expression of miRNAs in cancer cells is a widely accepted phenomenon, the molecular mechanisms underlying miR-3648 progression and metastasis in gastric cancer (GC) remain unclear. miR-3648 expression is downregulated and its ectopic expression in GC cells significantly suppressed cell proliferation and metastasis. Mechanistic analyses indicated that miR-3648 directly targets FRAT1 or FRAT2 and inhibits FRAT1- or FRAT2-mediated invasion and motility in vitro and in vivo. Moreover, FRAT1 physically interacted with FRAT2. Furthermore, FRAT1 overexpression promoted GC cell invasion, whereas siRNA-mediated repression of FRAT2 in FRAT1-overexpressing GC cells reversed its invasive potential. Besides, miR-3648 inactivated the Wnt/ß-catenin signalling pathway by downregulating FRAT1 and FRAT2 in GC. Interestingly, c-Myc, a downstream effector of Wnt/ß-catenin signalling, was also downregulated by miR-3648 overexpression. In turn, c-Myc negatively regulated miR-3648 expression by binding to the miR-3648 promoter. In addition, miR-3648 expression levels were negatively correlated with c-Myc, FRAT1, and FRAT2 expression in fresh gastric samples. Our studies suggest that miR-3648 acts as a tumour-suppressive miRNA and that the miR-3648/FRAT1-FRAT2/c-Myc negative feedback loop could be a critical regulator of GC progression.


Subject(s)
MicroRNAs , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , beta Catenin/genetics , beta Catenin/metabolism , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Feedback , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering , MicroRNAs/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Cell Movement/genetics
8.
Exp Cell Res ; 415(1): 113107, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35306026

ABSTRACT

The coiled-coil domain-containing protein 43 (CCDC43) is essential to promote gastric cancer (GC) proliferation and invasion, while four and a half LIM domains 1 (FHL1) involves GC cells apoptosis. We attempted to address inter-relationship between CCDC43 and FHL1 in modulating GC cells growth and apoptosis. Levels of protein expression were assessed by western blot, immunofluorescence. Using EdU, plate colony formation, Matrigel invasion and animal models, we evaluated the function in vitro and in vivo. Apoptosis was evaluated by flow cytometry and Hoechst 33258 staining. Reciprocal co-immunoprecipitation (co-IP) analyses indicated that CCDC43 physically interacted with FHL1. The expression of CCDC43 was negatively correlated with FHL1. Moreover, up-regulation of CCDC43 resulted in FHL1 level decline, and the reverse is also true. CCDC43 expressed jointly with FHL1 group significantly decreases the ability of the growth, metastasis and invasion of GC cells compared with that of the CCDC43 group. Furthermore, siRNA-mediated repression of CCDC43 results in dissociation from FHL1 and causes suppression of GC cell proliferation and metastasis. CCDC43 repression mediates the stability of FHL1 protein. In addition, CCDC43 interacts with FHL1. Knockdown of CCDC43 plus FHL1 overexpression inhibits proliferation and migration and induces apoptosis of GC cells in vitro and vivo.


Subject(s)
Stomach Neoplasms , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/pathology , Up-Regulation
9.
Sensors (Basel) ; 23(1)2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36616611

ABSTRACT

A multi-line structured light measurement method that combines a single-line and a three-line laser, in which precision sliding rails and displacement measurement equipment are not required, is proposed in this paper. During the measurement, the single-line structured light projects onto the surface of an object and the three-line structured light remains fixed. The single-line laser is moved and intersects with the three-line laser to form three intersection points. The single-line light plane can be solved using the camera coordinates of three intersection points, thus completing the real-time calibration of the scanned light plane. The single-line laser can be scanned at any angle to determine the overall complete three-dimensional (3D) shape of the object during the process. Experimental results show that this method overcomes the difficulty of obtaining information about certain angles and locations and can effectively recover the 3D shape of the object. The measurement system's repetition error is under 0.16 mm, which is sufficient to measure the 3D shapes of complicated workpieces.

10.
Cell Death Dis ; 13(1): 10, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930901

ABSTRACT

As an important regulator of intracellular protein degradation, the mechanism of the deubiquitinating enzyme family in tumour metastasis has received increasing attention. Our previous study revealed that USP3 promotes tumour progression and is highly expressed in gastric cancer (GC). Herein, we report two critical targets, COL9A3 and COL6A5, downstream of USP3, via the isobaric tags for relative and absolute quantification technique. Mechanistically, we observed that USP3 interacted with and stabilised COL9A3 and COL6A5 via deubiquitination in GC. Importantly, we found that COL9A3 and COL6A5 were essential mediators of USP3-modulated oncogenic activity in vitro and in vivo. Examination of clinical samples confirmed that elevated expression of USP3, concomitant with increased COL9A3 and COL6A5 abundance, correlates with human GC progression. These data suggest that USP3 promotes GC progression and metastasis by deubiquitinating COL9A3 and COL6A5. These findings identify a mechanism of GC metastasis regarding USP3-mediated deubiquitinating enzyme activity and suggest potential therapeutic targets for GC management.


Subject(s)
Carcinogenesis/metabolism , Collagen Type IX/metabolism , Collagen Type VI/metabolism , Lung Neoplasms/secondary , Signal Transduction/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Ubiquitin-Specific Proteases/metabolism , Ubiquitination/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Collagen Type IX/genetics , Collagen Type VI/genetics , Disease Models, Animal , Heterografts , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Transplantation/methods , RNA Interference , Transfection/methods , Ubiquitin-Specific Proteases/genetics
11.
Opt Express ; 29(20): 31324-31336, 2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34615227

ABSTRACT

In this paper, we have proposed and experimentally demonstrated a multiplexed sensing interrogation technique based on a flexibly switchable multi-passband RF filter with a polarization maintaining fiber (PMF) Solc-Sagnac loop. A high-order Solc-Sagnac loop can be used as a spectrum slicer as well as sensing heads, and a multi-passband microwave photonic filter (MPF) can be achieved together with a dispersive medium. Environmental parameter variations will cause a frequency shift of the corresponding passband of the MPF, so by employing only one Sagnac loop, it is possible to monitor several environmental parameters simultaneously. In this article, we have demonstrated and analyzed the performance of the flexibly switchable multi-passband MPF by using a second-order Solc-Sagnac loop. To demonstrate the temperature sensing capabilities of our interrogation system, we have applied temperature changes individually to Sensor Head 1 (L P M F 1 ≈0.97m) only, Sensor Head 2 (L P M F 2 ≈2.97m) only, and both Sensor Head 1 and 2 in the experiment. By monitoring frequency shift of the MPF's passbands, the sensitivities for Sensor Head 1 and Sensor Head 2 have been estimated to be -0.275 ± 0.011 MHz/℃ and -0.811 ± 0.013 MHz/℃ respectively, which show a good sensing linearity and stability. By utilizing the longer length of the sensing PMF, higher sensitivity can be achieved. By using Solc-Sagnac loop with higher order, more sensors can be multiplexed.

12.
Cancer Lett ; 521: 196-209, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34481934

ABSTRACT

Previous reports have shown that histone deacetylase inhibitors (HDACi) can alter miRNA expression in a range of cancers. Both the 5p-arm and 3p-arm of mature miRNAs can be expressed from the same precursor and involved in cancer progress. Nevertheless, the detailed mechanism by which vorinostat (SAHA), a HDACi, triggers miR-769-5p/miR-769-3p-mediated suppression of proliferation and induces apoptosis in gastric cancer (GC) cells remains elusive. Here, we showed that the miRNA-seq analysis of GC cells treated with SAHA identified seven differentially expressed miRNAs with both strands of the miRNA duplex. miR-769-5p/miR-769-3p expression was downregulated in GC tissues compared with normal tissues. Functionally, high expression of miR-769-5p/miR-769-3p blocked the malignant abilities of GC cells. Mechanistically, miR-769-5p/miR-769-3p targeted IGF1R and IGF1R overexpression rescued the effects of miR-769-5p/miR-769-3p on GC cells growth and metastasis. Moreover, STAT3 bound to the promoter of miR-769. Furthermore, miR-769-5p/miR-769-3p expression was negatively regulated by the STAT3-IGF1R-HDAC3 complex. Besides, miR-769-5p/miR-769-3p synergized with SAHA to promote GC cells apoptosis. Our studies suggest that miR-769-5p/miR-769-3p acts as a tumor suppressor by the STAT3-IGF1R-HDAC3 complex. Moreover, SAHA triggers miR-769-5p/miR-769-3p-mediated inhibition of proliferation and induces apoptosis in GC cells.

13.
Front Genet ; 12: 661306, 2021.
Article in English | MEDLINE | ID: mdl-34249086

ABSTRACT

BACKGROUND: It has been widely reported that epithelial-mesenchymal transition (EMT) is associated with malignant progression in gastric cancer (GC). Integration of the molecules related to EMT for predicting overall survival (OS) is meaningful for understanding the role of EMT in GC. Here, we aimed to establish an EMT-related gene signature in GC. METHODS: Transcriptional profiles and clinical data of GC were downloaded from The Cancer Genome Atlas (TCGA). We constructed EMT-related gene signature for predicting OS by using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. Time-dependent receiver operating characteristic (ROC), Kaplan-Meier analysis were performed to assess its predictive value. A nomogram combining the prognostic signature with clinical characteristics for OS prediction was established. And its predictive power was estimated by concordance index (C-index), time-dependent ROC curve, calibration curve and decision curve analysis (DCA). GSE62254 dataset from Gene Expression Omnibus (GEO) was used for external validation. Quantitative real-time PCR (qRT-PCR) was used to detected the mRNA expression of the five EMT-related genes in human normal gastric mucosal and GC cell lines. To further understand the potential mechanisms of the signature, Gene Set Enrichment Analysis (GSEA), pathway enrichment analysis, predictions of transcription factors (TFs)/miRNAs were performed. RESULTS: A novel EMT-related gene signature (including ITGAV, DAB2, SERPINE1, MATN3, PLOD2) was constructed for OS prediction of GC. With external validation, ROC curves indicated the signature's good performance. Patients stratified into high- and low-risk groups based on the signature yielded significantly different prognosis. Univariate and multivariate Cox regression suggested that the signature was an independent prognostic variable. Nomogram for prognostication including the signature presented better predictive accuracy and clinical usefulness than the similar model without risk score to some extent with external validation. The qRT-PCR assays suggested that high expression of the five EMT-related genes could be found in human GC cell lines compared with normal gastric mucosal cell line. GSEA and pathway enrichment analysis revealed that focal adhesion and ECM-receptor interaction might be the two important pathways to the signature. CONCLUSION: Our EMT-related gene signature may have practical application as an independent prognostic factor in GC.

14.
Aging (Albany NY) ; 13(12): 16043-16061, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34167089

ABSTRACT

HMGA1 protein is an architectural transcription factor that has been implicated in the progression of multiple malignant tumors. However, the role of HMGA1 in the growth and metastasis of gastric cancer (GC) has not yet been elucidated. Here, we show that HMGA1 is overexpressed in GC cells and the high expression of HMGA1 was correlated with worse survival in GC patients using a bioinformatics assay. Functionally, HMGA1 affected the EdU incorporation, colony formation, migration and invasion of GC cells by exogenously increasing or decreasing the expression of HMGA1. Mechanistically, HMGA1 directly bound to the SUZ12 and CCDC43 promoter and transactivated its expression in GC cells. Inhibition of SUZ12 and CCDC43 attenuated the proliferation, migration and invasiveness of HMGA1-overexpressing GC cells in vitro. Moreover, both HMGA1 and SUZ12/CCDC43 were highly expressed in cancer cells but not in normal gastric tissues, and their expressions were positively correlated. Finally, a tail vein metastatic assay showed that HMGA1 promoted SUZ12/CCDC43-mediated GC cell metastasis in vivo. Our findings suggest that HMGA1 promotes GC growth and metastasis by transactivating SUZ12 and CCDC43 expression, highlighting HMGA1 as a potential prognostic biomarker in the treatment of GC.


Subject(s)
HMGA1a Protein/metabolism , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/genetics , Transcriptional Activation/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Factors/metabolism , Up-Regulation/genetics
15.
Aging (Albany NY) ; 13(5): 6606-6624, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33535170

ABSTRACT

HOXA6 gene plays a role of the oncogene in various cancers. Nonetheless, its effect on gastric cancer (GC) occurrence and development is still unclear. We analysed whether HOXA6 interacts with the PBX2 protein using the STRING database. The molecular mechanism by which HOXA6 synergizes with PBX2 in GC metastasis is not fully understood. Here, we found that the expression of HOXA6 was increased in GC tissues and cell lines. The upregulation of HOXA6 was closely associated with differentiation, lymph node metastasis, AJCC stage, TNM stage, and poor survival outcome in GC patients based on tissue microarray (TMA) data. Moreover, the overexpression of HOXA6 promoted, whereas siRNA-mediated repression of HOXA6 inhibited, the cell proliferation, migration, and invasion of GC cells. Furthermore, HOXA6 could physically interact with and stabilize PBX2. In addition, HOXA6 and PBX2 expression was positively correlated in GC cells and tissue. HOXA6 and PBX2 suppression in GC cells also led to decreased migration and invasion potential in vitro. In vivo, HOXA6 was shown to cooperate with PBX2 to enhance cell metastasis via orthotopic implantation. These data indicate that HOXA6 promotes cell proliferation, migration, and invasion and that the HOXA6-PBX2 axis may be a useful biomarker for disease progression in GC.


Subject(s)
Homeodomain Proteins/metabolism , Lung Neoplasms/secondary , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Stomach Neoplasms/pathology , Animals , Cell Movement , Cell Proliferation , Female , Homeodomain Proteins/genetics , Humans , Male , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , Stomach Neoplasms/genetics
16.
Front Oncol ; 11: 767644, 2021.
Article in English | MEDLINE | ID: mdl-35004288

ABSTRACT

BACKGROUND: Accumulating studies have demonstrated the abnormal expressions and prognostic values of certain members of the tripartite motif (TRIM) family in diverse cancers. However, comprehensive prognostic values of the TRIM family in hepatocellular carcinoma (HCC) are yet to be clearly defined. METHODS: The prognostic values of the TRIM family were evaluated by survival analysis and univariate Cox regression analysis based on gene expression data and clinical data of HCC from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The expression profiles, protein-protein interaction among the TRIM family, prediction of transcription factors (TFs) or miRNAs, genetic alterations, correlations with the hallmarks of cancer and immune infiltrates, and pathway enrichment analysis were explored by multiple public databases. Further, a TRIM family gene-based signature for predicting overall survival (OS) in HCC was built by using the least absolute shrinkage and selection operator (LASSO) regression. TCGA-Liver Hepatocellular Carcinoma (LIHC) cohort was used as the training set, and GSE76427 was used for external validation. Time-dependent receiver operating characteristic (ROC) and survival analysis were used to estimate the signature. Finally, a nomogram combining the TRIM family risk score and clinical parameters was established. RESULTS: High expressions of TRIM family members including TRIM3, TRIM5, MID1, TRIM21, TRIM27, TRIM32, TRIM44, TRIM47, and TRIM72 were significantly associated with HCC patients' poor OS. A novel TRIM family gene-based signature (including TRIM5, MID1, TRIM21, TRIM32, TRIM44, and TRIM47) was built for OS prediction in HCC. ROC curves suggested the signature's good performance in OS prediction. HCC patients in the high-risk group had poorer OS than the low-risk patients based on the signature. A nomogram integrating the TRIM family risk score, age, and TNM stage was established. The ROC curves suggested that the signature presented better discrimination than the similar model without the TRIM family risk score. CONCLUSION: Our study identified the potential application values of the TRIM family for outcome prediction in HCC.

17.
Cell Oncol (Dordr) ; 43(6): 1017-1033, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32857323

ABSTRACT

PURPOSE: Growing evidence indicates that aberrant expression of microRNAs contributes to tumor development. However, the biological role of microRNA-4490 (miR-4490) in gastric cancer (GC) remains to be clarified. METHODS: To explore the function of miR-4490 in GC, we performed colony formation, EdU incorporation, qRT-PCR, Western blotting, in situ hybridization (ISH), immunohistochemistry (IHC), flow cytometry, ChIP and dual-luciferase reporter assays. In addition, the growth, migration and invasion capacities of GC cells were evaluated. RESULTS: We found that miR-4490 was significantly downregulated in primary GC samples and in GC-derived cell lines compared with normal controls, and that this expression level was negatively correlated with GC malignancy. Exogenous miR-4490 expression not only reduced cell cycle progression and proliferation, but also significantly inhibited GC cell migration, invasion and epithelial-mesenchymal transition (EMT) in vitro. Mechanistically, we found that miR-4490 directly targets USP22, which mediates inhibition of GC cell proliferation and EMT-induced metastasis in vitro and in vivo. Moreover, we found through luciferase and ChIP assays that transcription factor POU2F1 can directly bind to POU2F1 binding sites within the miR-4490 and USP22 promoters and, by doing so, modulate their transcription. Spearman's correlation analysis revealed a positive correlation between USP22 and POU2F1 expression and negative correlations between miR-4490 and USP22 as well as miR-4490 and POU2F1 expression in primary GC tissues. CONCLUSION: Based on our results we conclude that miR-4490 acts as a tumor suppressor, and that the POU2F1/miR-4490/USP22 axis plays an important role in the regulation of growth, invasion and EMT of GC cells.


Subject(s)
MicroRNAs/metabolism , Octamer Transcription Factor-1/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Octamer Transcription Factor-1/genetics , Promoter Regions, Genetic/genetics , Signal Transduction , Transcriptional Activation/genetics , Ubiquitin Thiolesterase/genetics
18.
Int J Oncol ; 56(6): 1499-1508, 2020 06.
Article in English | MEDLINE | ID: mdl-32236592

ABSTRACT

The development of malignant tumors is a series of complex processes, the majority of which have not been elucidated. The aim of the present study was to investigate the microRNAs (miRNAs/miR) that affect the migration and invasion abilities of CRC cells. Our previous reports have revealed that miR­500a­5p suppressed CRC cell growth and malignant transformation. The present study demonstrated that overexpression of miR­500a­5p reduced the expression of vimentin, while increasing the expression of E­cadherin. Inhibition of miR­500a­5p resulted in spindle­like morphological changes and reorganization of F­actin in CRC cells. Furthermore, miR­500a­5p attenuated the transforming growth factor­ß signaling pathway in EMT. Additionally, emodin inhibited the miR­500a­5p inhibitor and suppressed the EMT process. In animal models of metastasis using nude mice, EMT and LoVo cell metastasis was modulated by miR­500a­5p. Therefore, the findings of the present study demonstrated that miR­500a­5p is associated with a positive therapeutic outcome in terms of invasion/migration of CRC cells and mesenchymal­like cell changes.


Subject(s)
Colorectal Neoplasms/pathology , Down-Regulation , MicroRNAs/genetics , Signal Transduction , Actins/genetics , Actins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vimentin/genetics , Vimentin/metabolism
19.
Cancer Med ; 9(11): 3932-3943, 2020 06.
Article in English | MEDLINE | ID: mdl-32281284

ABSTRACT

BACKGROUND: HOXD9, a Hox family member, is involved in cancer growth and metastasis. But, its regulation mechanism at the molecular level particularly in colorectal cancer (CRC), is mostly unknown. METHODS: The HOXD9 protein expression levels were analyzed using immunofluorescence, immunohistochemistry (IHC) assays, and western blot. The in vivo and in vitro roles of HOXD9 in CRC were determined using colony formation and EdU incorporation, CCK-8, wound scratch and transwell invasion assay, and animal models. RESULTS: Expression of HOXD9 was higher in CRC than in matched healthy tissues. High expression of HOXD9 has significantly associated with the American Joint Committee on Cancer (AJCC) stages, tumor differentiation, lymph node metastasis, and other serious invasions, and it had a poor prognosis. In vitro, HOXD9 encouraged proliferation, movement and EMT processes in cells of CRC. Also, TGF-ß1 promoted the expression of HOXD9 and this effect was dependent on the dose and downregulation of HOXD9 repressed TGF-ß1 -induced EMT. In vivo, HOXD9 promoted the invasive and metastasis of CRC cells via orthotopic implantation. CONCLUSIONS: The ectopic expression of HOXD9 promoted the invasion metastasis in cells of the colorectal tumor by induction of EMT in vitro and vivo.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Prognosis , Survival Rate , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Cancer Lett ; 482: 90-101, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32278016

ABSTRACT

Previous studies have shown an association between coiled-coil domain-containing (CCDC) genes and different cancers. Our previous studies revealed that CCDC43 is highly expressed in colorectal cancer, but the expression and molecular mechanisms of CCDC43 in gastric cancer (GC) are yet to be determined. Here, we show that CCDC43 is overexpressed in gastric tissues. CCDC43 expression is closely related to tumor differentiation, lymph-node-metastasis, and prognosis of gastric cancer. Overexpression of CCDC43 promotes the proliferation, invasion, and metastasis of GC cells. CCDC43 may upregulate and stabilize ADRM1, resulting in the construction of the ubiquitin-mediated proteasome. In contrast, inhibition of ADRM1 could reverse the function of CCDC43 in GC both in vitro and in vivo. Our data demonstrate that transcription factor YY1 directly binds to CCDC43 and ADRM1 gene promoters, leading to over-expression of CCDC43 and ADRM1. Furthermore, in vitro experiments demonstrate that knock down of CCDC43 or ADRM1 attenuates the YY1-mediated malignant phenotypes. Finally, the association among YY1, CCDC43 and ADRM1 is validated in clinical samples. Our findings suggest that the CCDC43-ADRM1 axis regulated by YY1, promotes proliferation and metastasis of GC, and the axis may be a potential therapeutic target for GC.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Up-Regulation , YY1 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Mice , Neoplasm Transplantation , Prognosis , Stomach Neoplasms/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...