Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cancers (Basel) ; 14(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36428666

ABSTRACT

The synergistic effect of combining immune checkpoint inhibitors (ICIs) with neoadjuvant chemo(radio)therapy (nCRT) in colorectal cancer is still limited. We aimed to understand the impact of nCRT on the tumor microenvironment and to explore favorable immune markers of this combination. Herein, we investigated the expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), CD86, CD4, and CD8 after nCRT and its association with clinicopathological characteristics. Immunostaining of immune-related molecules was performed in 255 surgically resected specimens from rectal cancer patients treated with nCRT. CD4 and CD8 expression on the tumor (tCD4/CD8), stroma (sCD4/CD8), and invasive front (iCD4/CD8) was evaluated. The expression levels of immune-related molecules were significantly lower in the nCRT-treated group, except for CTLA-4 and sCD8. However, patients with higher sCD8+ cell density and CTLA-4 expression had better progression-free survival (PFS) and distant metastasis-free survival (DMFS). In addition, higher CD86 expression was associated with poorer overall survival (OS). Higher CTLA-4 expression was associated with higher tCD8+ cell density, whereas CD86 expression was correlated with the cell density of t/sCD8. Prognostic analysis confirmed that the relationships between CTLA-4 and DMFS as well as CD86 and OS were significantly correlated in low rather than high CD8+ cell density. Further the combination of CD8+ cell density and CD86 expression was shown to be an independent prognostic factor of OS, whereas the combination of CTLA-4 was not for DMFS. Together, these results demonstrate significant correlations between CD86 expression and t/sCD8+ cell density in rectal cancer after nCRT and could potentially have clinical implications for combining ICIs and nCRT.

2.
BMC Cancer ; 22(1): 928, 2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36030223

ABSTRACT

INTRODUCTION: Neuroendocrine differentiation (NED) in colorectal cancer (CRC) cells has been known for decades, and our previous meta-analysis indicated that CRC patients with neuroendocrine differentiation have a lower 5-year survival rate. In recent years, an increasing number of studies have found that exosome-derived long non-coding RNAs (lncRNAs) play important roles in cancer progression and metastasis. However, the functions and mechanism of exosome-derived lncRNAs in CRC with neuroendocrine differentiation are not yet fully clear. MATERIALS AND METHODS: The clinical significance of NED was assessed in a retrospective study of 105 patients. Next-generation sequencing and bioinformatics analysis were conducted to select lnc-HOXB8-1:2 for further study. Using immunohistochemistry, qRT-PCR, western blot, transwell assay, immunofluorescence assay, fluorescence in situ hybridization assay and dual-luciferase reporter assay, the oncogenic role of exosome-derived lnc-HOXB8-1:2 was determined in CRC with NED. The mechanism underlying the lnc-HOXB8-1:2/hsa-miR-6825-5p/CXCR3 axis was also explored. RESULTS: NED was a risk factor for the progression and mortality of CRC. lnc-HOXB8-1:2, derived from exosomes secreted by neuroendocrine differentiated colon cancer cells, was identified in our study. The proportion of M2 macrophages and the migration and invasion capacities of tumor-associated macrophages (TAMs) markedly increased after the addition of neuroendocrine differentiated CRC cell-derived exosomes. More excitingly, the expression of lnc-HOXB8-1:2 and the protein level of CXCR3 were also upregulated in TAMs. The lnc-HOXB8-1:2/hsa-miR-6825-5p/CXCR3 axis was predicted via miRanda software and confirmed by the dual-luciferase reporter assay. Furthermore, the increased expression of lnc-HOXB8-1:2 was accompanied by downregulation of hsa-miR-6825-5p expression and upregulation of CXCR3 protein levels. Overexpression of hsa-miR-6825-5p also reduced CXCR3 expression. CONCLUSION: lnc-HOXB8-1:2 in exosomes derived from neuroendocrine differentiated CRC cells acted as a ceRNA competitively binding hsa-miR-6825-5p to upregulate CXCR3 expression and leading to TAM infiltration and M2 polarization, which promotes neuroendocrine differentiated CRC progression.


Subject(s)
Colorectal Neoplasms , Exosomes , MicroRNAs , RNA, Long Noncoding , Tumor-Associated Macrophages , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Homeodomain Proteins , Humans , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Retrospective Studies , Tumor-Associated Macrophages/cytology
3.
BMC Gastroenterol ; 22(1): 87, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35227194

ABSTRACT

BACKGROUND: Abnormal hypermethylation of the septin 9 gene was an inchoate incident in some cancers. Though latest several researches had paid attention to its value in prognosis, the consequences were not distinctly, especially in colorectal cancer (CRC) with stage II and stage III. PURPOSE: The aim of this research was to pick up the prognostic value of the methylated septin 9 gene (mSEPT9) in CRC patients, particularly in TNM stage II-III. METHODS: Blood samples before surgery were obtained from 144 CRC patients, of which there were 94 with stage II and stage III. mSEPT9 was considered positive when the cycle number of the peak reaction (Ct) was lower than the threshold value (41.0) for two times during three times PCR test. mSEPT9 and other relative factors of prognosis were estimated by survival analysis. The level of septin 9 in tissues was tested by immunohistochemical (IHC). RESULTS: Stage II and stage III patients with mSEPT9 positive (mSEPT9+) had a lower disease-free survival (DFS) rate than those with mSEPT9 negative (mSEPT9-) (2-year DFS rates, 52.1% vs 73.9%, P = 0.014). In multivariate regression analysis, mSEPT9 was also an independent predictor of prognosis (HR = 2.741, P = 0.009). The risk of local recurrence or distant metastasis in CRC patients after surgery was mSEPT9+ with stage III, mSEPT9- with stage III/mSEPT9+ with stage II, and mSEPT9- with stage II (P = 0.001), from highest to lowest. In addition, mSEPT9 was strongly associated with TNM staging, tumor immersion depth, distant metastasis, differentiation degree, vascular invasion and microsatellite. When we explored the associations between septin 9 protein level revealed by IHC and other elements, recurrence/progression (R = - 0.523, P = 0.001), mSEPT9 status (R = - 0.451, P = 0.004) and T stage (R = - 0.375, P = 0.017) showed significant correlations. CONCLUSIONS: Positive mSEPT9 is a poor prognostic marker for CRC patients in stage II and III. It is also a powerful complement to TNM staging in predicting postoperative DFS of CRC patients of stage II and III.


Subject(s)
Colorectal Neoplasms , Septins , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , DNA Methylation , Humans , Male , Neoplasm Staging , Prognosis , Septins/genetics , Septins/metabolism
4.
J Hematol Oncol ; 14(1): 194, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34781990

ABSTRACT

BACKGROUND: The main cause of death in colorectal cancer patients is metastasis. Accumulating evidences suggest that circRNA plays pivotal roles in cancer initiation and development. However, the underlying molecular mechanisms of circRNAs that orchestrate cancer metastasis remain vague and need further clarification. METHODS: Two paired CRC and adjacent normal tissues were used to screen the upregulated circRNAs by circRNA-seq; then, cell invasion assay was applied to confirm the functional invasion-related circRNAs. According to the above methods, circHERC4 (hsa_circ_0007113) was selected for further research. Next, we investigated the clinical significance of circHERC4 in a large cohort of patients with CRC. The oncogenic activity of circHERC4 was investigated in both CRC cell lines and animal xenograft studies. Finally, we explored the molecular mechanisms underlying circHERC4 as a malignant driver. RESULTS: We demonstrated that circHERC4 was aberrantly elevated in CRC tissues (P < 0.001), and was positively associated with lymph node metastasis and advanced tumor grade (P < 0.01). Notably, the expression of circHERC4 was associated with worse survival in patients with CRC. Silencing of circHERC4 significantly inhibited the proliferation and migration of two highly aggressive CRC cell lines and reduced liver and lung metastasis in vivo. Mechanistically, we revealed that circHERC4 inactivated the tumor suppressor, miR-556-5p, leading to the activation of CTBP2/E-cadherin pathway which promotes tumor metastasis in CRC. CONCLUSIONS: CircHERC4 exerts critical roles in promoting tumor aggressiveness through miR-556-5p/CTBP2/E-cadherin pathway and is a prognostic biomarker of the disease, suggesting that circHERC4 may serve as an exploitable therapeutic target for patients with CRC.


Subject(s)
Alcohol Oxidoreductases/genetics , Antigens, CD/genetics , Cadherins/genetics , Co-Repressor Proteins/genetics , Colorectal Neoplasms/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice, Inbred BALB C , Neoplasm Invasiveness/genetics
5.
BMC Cancer ; 21(1): 718, 2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34147083

ABSTRACT

BACKGROUND: Liver metastasis is the most common cause of death in patients with colorectal cancer (CRC). Phosphatase of regenerating liver-3 induces CRC metastasis by epithelial-to-mesenchymal transition, which promotes CRC cell liver metastasis. Mesenchymal-to-epithelial transition (MET), the opposite of epithelial-to-mesenchymal transition, has been proposed as a mechanism for the establishment of metastatic neoplasms. However, the molecular mechanism of MET remains unclear. METHODS: Using Immunohistochemistry, western blotting, invasion assays, real-time quantitative PCR, chromatin immunoprecipitation, luciferase reporter assays, human miRNA arrays, and xenograft mouse model, we determined the role of hepatocyte exosome-derived miR-203a-3p in CRC MET. RESULTS: In our study, we found that miR-203a-3p derived from hepatocyte exosomes increased colorectal cancer cells E-cadherin expression, inhibited Src expression, and reduced activity. In this way miR-203a-3p induced the decreased invasion rate of CRC cells. COCLUSION: MiR-203a-3p derived from hepatocyte exosomes plays an important role of CRC cells to colonize in liver.


Subject(s)
Colorectal Neoplasms/genetics , Exosomes/genetics , MicroRNAs/genetics , Animals , Cell Proliferation , Colorectal Neoplasms/pathology , Congenital Hypothyroidism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Humans , Mice , Mice, Nude , Thyroid Dysgenesis
6.
BMC Microbiol ; 21(1): 156, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34044781

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common cancers. In recent studies, the gut microbiota has been reported to be potentially involved in aggravating or favoring CRC development. However, little is known about the microbiota composition in CRC patients after treatment. In this study, we explored the fecal microbiota composition to obtain a periscopic view of gut microbial communities. We analyzed microbial 16S rRNA genes from 107 fecal samples of Chinese individuals from three groups, including 33 normal controls (NC), 38 CRC patients (Fa), and 36 CRC post-surgery patients (Fb). RESULTS: Species richness and diversity were decreased in the Fa and Fb groups compared with that of the NC group. Partial least squares discrimination analysis showed clustering of samples according to disease with an obvious separation between the Fa and NC, and Fb and NC groups, as well as a partial separation between the Fa and Fb groups. Based on linear discriminant analysis effect size analysis and a receiver operating characteristic model, Fusobacterium was suggested as a potential biomarker for CRC screening. Additionally, we found that surgery greatly reduced the bacterial diversity of microbiota in CRC patients. Some commensal beneficial bacteria of the intestinal canal, such as Faecalibacterium and Prevotella, were decreased, whereas the drug-resistant Enterococcus was visibly increased in CRC post-surgery group. Meanwhile, we observed a declining tendency of Fusobacterium in the majority of follow-up CRC patients who were still alive approximately 3 y after surgery. We also observed that beneficial bacteria dramatically decreased in CRC patients that recidivated or died after surgery. This revealed that important bacteria might be associated with prognosis. CONCLUSIONS: The fecal bacterial diversity was diminished in CRC patients compared with that in NC. Enrichment and depletion of several bacterial strains associated with carcinomas and inflammation were detected in CRC samples. Fusobacterium might be a potential biomarker for early screening of CRC in Chinese or Asian populations. In summary, this study indicated that fecal microbiome-based approaches could be a feasible method for detecting CRC and monitoring prognosis post-surgery.


Subject(s)
Bacteria/isolation & purification , Colorectal Neoplasms/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Aged , Bacteria/classification , Bacteria/genetics , Biodiversity , Colorectal Neoplasms/surgery , Female , Humans , Male , Middle Aged
7.
J Gastrointest Oncol ; 12(6): 2749-2762, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35070404

ABSTRACT

BACKGROUND: Though colon cancer (CC) is one of the most malignant tumors across the world, CC patients with microsatellite instability-high (MSI-H) in stage II seem to have a better prognosis. However, the molecular mechanisms underlying the phenomena haven't been elucidated yet. METHODS: This study enrolled 322 CCs with known microsatellite status from GSE143985, GSE39582 and GSE92921 in the Gene Expression Omnibus (GEO) database. Robust rank aggregation (RRA) analysis, univariate Cox regression analysis and multivariate Cox stepwise regression analysis were performed to identify genes and construct risk score signature. Kaplan-Meier and receiver operating characteristic (ROC) curves analyses were used to evaluate the prognostic value of the signature. The potential mechanisms underlying this signature were assessed in the Metascape database, gene set enrichment analysis (GSEA) and immune infiltration analysis. RESULTS: RRA analysis identified 40 differently expressed genes (DEGs). A 3-gene risk score signature (MKQ signature) associated with disease-free survival (DFS) was generated. DFS was significantly longer in CC patients with lower than higher scores (P=0.0046). The areas under curves (AUCs) of the time-dependent ROC curves of MKQ signature at 1-, 3- and 5-year DFS were 1, 0.963 and 0.961 respectively. Recurrence-free survival (RFS) was significantly longer in patients in GSE39582 with lower than higher risk scores (P=0.032). The AUCs for 1-, 3- and 5-year RFS in GSE39582 were 0.63, 0.618 and 0.583, respectively, validating the value of the MKQ signature. Functional annotation and GSEA revealed that the MKQ signature was associated with multiple immune-related pathways. Immune cell infiltration was found to differ in patients differing in the MKQ signature. CONCLUSIONS: Gene expression and microsatellite status identified a 3-gene signature (MKQ signature) that could facilitate risk-stratified management in patients with stage II CC. Dysregulation of MSMB, KRT23, and QPRT can serve as prognostic markers in stage II CC.

8.
Clin Transl Gastroenterol ; 11(12): e00265, 2020 12.
Article in English | MEDLINE | ID: mdl-33512811

ABSTRACT

INTRODUCTION: Circulating tumor cells (CTCs) and phosphatase of regenerating liver-3 (PRL-3) have been considered to be significant prognostic indicators in metastatic colorectal cancer (CRC). This study discusses the prognostic significance of mesenchymal CTCs with PRL-3 (M+ PRL-3+ CTCs) in postoperative patients with CRC. METHODS: We detected CTC subtypes (including epithelial CTCs, biphenotypic epithelial/mesenchymal CTCs, and mesenchymal CTCs) and PRL-3 in CTCs from the peripheral blood samples of 156 patients. Receiver operating characteristic curve analysis, Kaplan-Meier analysis, and Cox proportional hazards regression analysis were performed to identify the prognostic value of mesenchymal CTCs with PRL-3+. Immunohistochemistry was used to detect the expression of PRL-3 in tumor tissues from some of the patients to explore the connection between CTCs and tissues. RESULTS: All CTCs were positive in all samples, both mesenchymal CTCs and PRL-3-positive cells. The count of mesenchymal and PRL-3+ CTCs was significantly associated with recurrence, and the optimal cutoff value was 2 (area under the curve = 0.690, P < 0.001). In addition, these patients had a significantly shorter median disease-free survival than those who did not fulfill the criteria (8.5 vs 24 months, P < 0.001) according to multivariable and multinomial logistic regression. Immunohistochemistry was applied to explore the associations between PRL-3 expression and significant prognostic risk factors, including recurrence (R = 0.566; P < 0.001), and M+ PRL-3+ status in CTCs (R = 0.452; P = 0.001). DISCUSSION: The status of M+ PRL-3+ in CTCs may serve as a crucial prognostic marker for assessing clinical outcomes in CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/mortality , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/epidemiology , Neoplastic Cells, Circulating/metabolism , Protein Tyrosine Phosphatases/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Colectomy , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Proctectomy , Prognosis , Prospective Studies , Protein Tyrosine Phosphatases/analysis , ROC Curve , Risk Assessment/methods , Young Adult
10.
Oncol Rep ; 41(5): 2790-2802, 2019 May.
Article in English | MEDLINE | ID: mdl-30864736

ABSTRACT

Protein phosphatase of regenerating liver­3 (PRL­3) is considered to be metastasis­associated phosphatase and is associated with a poor prognosis. Additionally, tumor­associated macrophages (TAMs) participate in cancer progression. A previous study demonstrated that PRL­3 promotes invasion and metastasis by inducing TAM infiltration. However, the underlying mechanism has not been elucidated. In the present study, western blot analysis, polymerase chain reaction, immunohistochemistry, ELISA, mouse model experiments and functional experiments were performed to confirm that the interaction between TAMs and colorectal cancer (CRC) cells induced epithelial­mesenchymal transition (EMT)­associated features in CRC cells by activating mitogen­activated protein kinase (MAPK) pathways in TAMs and upregulating the expression of interleukin (IL)­6 and IL­8. The neutralization of IL­6 and IL­8 reduced EMT and the invasive and migratory abilities of CRC cells. Therefore, IL­6 and IL­8 were considered important factors in EMT, and in CRC invasion and metastasis. In addition, increased angiogenesis was observed after TAMs were co­cultured with CRC cells that overexpress PRL­3. Vascular endothelial growth factor­A was significantly upregulated, and the nuclear factor­κB (NF­κB) signaling pathway was activated in CRC cells after co­culture. Moreover, nude mice injected with CRC cells with high PRL­3 expression levels tended to generate larger xenografts. Immunohistochemistry results from xenografted CRC cells overexpressing PRL­3 also confirmed the activation of MAPK pathways in xenografts. Overall, the findings indicate that PRL­3 promotes CRC cell invasion and metastasis by activating MAPK pathways in TAMs to initiate the EMT, and PRL­3 promotes angiogenesis by activating the NF­κB pathway in CRC cells.


Subject(s)
Colorectal Neoplasms/pathology , Macrophages/immunology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/pathology , Protein Tyrosine Phosphatases/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/immunology , Epithelial-Mesenchymal Transition/immunology , Humans , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Proteins/immunology , Neovascularization, Pathologic/immunology , Protein Tyrosine Phosphatases/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
11.
J Cancer Res Ther ; 14(4): 772-779, 2018.
Article in English | MEDLINE | ID: mdl-29970651

ABSTRACT

AIM: The aim of this is study is to assess the efficacy and safety of conversion capecitabine plus oxaliplatin (XELOX) in Chinese patients with potentially resectable colorectal liver metastases (CLMs). PATIENTS AND METHODS: Thirty patients (median age 57.5 years) with potentially resectable CLMs were treated with XELOX in a single-arm, open-label, nonrandomized, multicenter clinical trial. RESULTS: The objective response rate in the 30 patients was 40% (95% confidence interval: 22.7%-59.4%), and the rate of conversion to resectable CLMs was 43.3%. Patients who underwent liver resection (n = 11) had a longer median progression-free survival and overall survival than those who did not. XELOX showed an acceptable safety profile. CONCLUSION: XELOX may effectively convert potentially resectable CLM into resectable CLM, providing survival benefits with a favorable safety profile. CLINICAL TRIALS.GOV IDENTIFIER: NCT 00997685.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine/administration & dosage , Female , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Treatment Outcome
12.
Mol Cancer Ther ; 17(1): 276-289, 2018 01.
Article in English | MEDLINE | ID: mdl-29051319

ABSTRACT

Both phosphatase of regenerating liver-3 (PRL-3) and tumor-associated macrophages (TAM) influence cancer progression. Whether PRL-3 plays a critical role in colorectal cancer invasion and metastasis by inducing TAM infiltration remains unclear. In the current study, we investigated the effects of chemokine ligand 26 (CCL26) on TAM infiltration and colorectal cancer invasion and the underlying mechanism in colorectal cancer cells by overexpressing or silencing PRL-3. We found that PRL-3 upregulated CCL26 expression correlatively and participated in cell migration, according to the results of gene ontology analysis. In addition, IHC analysis results indicated that the PRL-3 and CCL26 levels were positively correlated and elevated in stage III and IV colorectal cancer tissues and were associated with a worse prognosis in colorectal cancer patients. Furthermore, we demonstrated that CCL26 induced TAM infiltration by CCL26 binding to the CCR3 receptor. When LoVo-P and HT29-C cells were cocultured with TAMs, CCL26 binding to the CCR3 receptor enhanced the invasiveness of LoVo-P and HT29-C cells by mobilizing intracellular Ca2+of TAMs to increase the expression of IL6 and IL8. In addition, IHC results indicated that protein levels of CCR3 and TAMs counts were higher in stage III and IV colorectal cancer tissues and correlated with CCL26. Moreover, similar results were observed in vivo using mice injected with LoVo-P and HT29-C cells. These data indicate that PRL-3 may represent a potential prognostic marker that promotes colorectal cancer invasion and metastasis by upregulating CCL26 to induce TAM infiltration. Mol Cancer Ther; 17(1); 276-89. ©2017 AACR.


Subject(s)
Chemokine CCL26/immunology , Colorectal Neoplasms/immunology , Macrophages/immunology , Neoplasm Proteins/immunology , Protein Tyrosine Phosphatases/immunology , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , HT29 Cells , Humans , Macrophages/pathology , Male , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Transfection
13.
Int J Oncol ; 51(4): 1271-1279, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28791350

ABSTRACT

Phosphatase of regenerating liver-3 (PRL-3) has been found to be overexpressed in liver metastases of colorectal cancer and rarely expressed in primary tumors, which plays an important role in the metastasis of colorectal cancer cells. Metabolism reprogramming has been found to be a hallmark of cancer cells, and aerobic glycolysis is a metabolic adaption for cancer cells and promotes cell proliferation. However, the association between PRL-3 and glycolysis in colorectal cancer cells is not well understood. In the present study, we explored the association between PRL-3 and glycolysis. We found that PRL-3 improved colorectal cancer cell glucose assumption, lactate production and reduced intracellular ROS levels. Besides, PRL-3 improved the expression of Glut1, HK2, PKM2 and LDHA, which are important glycolysis related molecules and enzymes. Moreover, we explored IL-8 mediated enhancement of glycolysis by PRL-3. More importantly, the proliferation and invasion of colorectal cancer cells were enhanced significantly by PRL-3 through improving glycolysis. Taken together, these results implicated the important role of PRL-3 in glycolysis metabolism through improving IL-8 secretion in colorectal cancer cells, and PRL-3 mediated glycolysis contributed to the promotion of cancer metastasis.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Interleukin-8/metabolism , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Cellular Reprogramming/physiology , Glycolysis , Humans , Neoplasm Invasiveness , Reactive Oxygen Species/metabolism
14.
PLoS One ; 11(8): e0159954, 2016.
Article in English | MEDLINE | ID: mdl-27532268

ABSTRACT

Dual-specificity tyrosine-phosphorylation-regulated kinase 2 (DYRK2) is a member of dual-specificity kinase family, which could phosphorylate both Ser/Thr and Tyr substrates. The role of DYRK2 in human cancer remains controversial. For example, overexpression of DYRK2 predicts a better survival in human non-small cell lung cancer. In contrast, amplification of DYRK2 gene occurs in esophageal/lung adenocarcinoma, implying the role of DYRK2 as a potential oncogene. However, its clinical role in colorectal cancer (CRC) has not been explored. In this study, we analyzed the expression of DYRK2 from Oncomine database and found that DYRK2 level is lower in primary or metastatic CRC compared to adjacent normal colon tissue or non-metastatic CRC, respectively, in 6 colorectal carcinoma data sets. The correlation between DYRK2 expression and clinical outcome in 181 CRC patients was also investigated by real-time PCR and IHC. DYRK2 expression was significantly down-regulated in colorectal cancer tissues compared with adjacent non-tumorous tissues. Functional studies confirmed that DYRK2 inhibited cell invasion and migration in both HCT116 and SW480 cells and functioned as a tumor suppressor in CRC cells. Furthermore, the lower DYRK2 levels were correlated with tumor sites (P = 0.023), advanced clinical stages (P = 0.006) and shorter survival in the advanced clinical stages. Univariate and multivariate analyses indicated that DYRK2 expression was an independent prognostic factor (P < 0.001). Taking all, we concluded that DYRK2 a novel prognostic biomarker of human colorectal cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Biomarkers, Tumor/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/mortality , Databases, Genetic , Disease-Free Survival , Female , HCT116 Cells , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Treatment Outcome , Dyrk Kinases
15.
Oncotarget ; 7(19): 27394-407, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27034164

ABSTRACT

Our previous study revealed that neuroendocrine differentiation in colorectal cancer is one of the important factors leading to worse prognosis. In this study, we apply immunohistochemical staining, Western-blot, RT-PCR and ELISA to investigate the underlying mechanism that how the neuroendocrine differentiation to affect the prognosis of colorectal cancer. The interaction of colorectal cancer cells, neuroendocrine-like cells and tumor-associated macrophages in colorectal cancer progress is also investigated. By analyzing 82 cases of colorectal cancer patients treated in our institution, we found that colorectal adenocarcinoma with neuroendocrine differentiation had increasing number of tumor-associated macrophages and worse prognosis. Further evaluation of cytology showed that neuroendocrine cells have the ability to recruit tumor-associated macrophages to infiltrate the tumor tissue, and the tumor-associated macrophages enhance the proliferation and invasion abilities of the colon cancer cells. Moreover, we confirmed that CXCL10 and CXCL11 are the key chemokines in neuroendocrine-like cells and they promote the chemotaxis activity of tumor-associated macrophages. The secretion of CXCL10 and CXCL11 by neuroendocrine-like cells can recruit tumor-associated macrophages to infiltrate in tumor tissues. The latter enhances the proliferation and invasion of colorectal cancer cell and lead to poor prognosis.


Subject(s)
Adenocarcinoma/metabolism , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Colorectal Neoplasms/metabolism , Macrophages/metabolism , Neuroendocrine Cells/metabolism , Adenocarcinoma/pathology , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Chemotaxis , Colorectal Neoplasms/pathology , Female , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , HT29 Cells , Humans , Male , Middle Aged , Neuroendocrine Cells/pathology , Prognosis
16.
Tumour Biol ; 37(8): 11299-309, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26951514

ABSTRACT

It is critical to understand the pathogenesis of preinvasive stages of pancreatic duct adenocarcinoma (PDAC) for developing novel potential diagnostic and therapeutic targets. The polycomb group family member B-lymphoma Moloney murine leukemia virus insertion region-1 (Bmi1) is overexpressed and involved in cancer progression in PDAC; however, its role in the multistep malignant transformation of human pancreatic duct cells has not been directly demonstrated. In this study, we stably expressed Bmi1 in a model of telomerase-immortalized human pancreatic duct-derived cells (HPNE) and showed that Bmi1 promoted HPNE cell proliferation, migration, and invasion but not malignant transformation. We then used mutant KRASG12D as a second oncogene to transform HPNE cells and showed that it further enhanced Bmi1-induced malignant potential. More importantly, coexpression of KRASG12D and Bmi1 caused anchorage-independent growth transformation in vitro but still failed to produce tumors in nude mice. Finally, we found that mutant KRASG12D induced HPNE-Bmi1 cells to undergo partial epithelial-mesenchymal transition (EMT) likely via upregulation of snail. Knockdown of KRASG12D significantly reduced the expression of snail and vimentin at both the messenger RNA (mRNA) and protein level and further impaired the anchorage-independent growth capability of invasive cells. In summary, our findings demonstrate that coexpression of Bmi1 and KRASG12D could lead to transformation of HPNE cells in vitro and suggest potential new targets for diagnosis and treatment of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Heterografts , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Int J Mol Sci ; 16(8): 19401-18, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26287184

ABSTRACT

The potential anti-neoplastic activity of terpenoids is of continued interest. In this study, we investigate whether methyl sartortuoate, a terpenoid isolated from soft coral, induced cell cycle arrest and apoptosis in a human colon cancer cell line. Culture studies found that methyl sartortuoate inhibited colon cancer cell (LoVo and RKO) growth and caused apoptotic death in a concentration- and time-dependent manner, by activation of caspase-8, caspase-9, caspase-3, p53 and Bax, and inactivation of B-cell lymphoma 2 (Bcl-2) apoptosis regulating proteins. Methyl sartortuoate treatment led to reduced expression of cdc2 and up-regulated p21 and p53, suggesting that Methyl sartortuoate induced G2-M arrest through modulation of p53/p21/cdc2 pathways. Methyl sartortuoate also up-regulated phospho-JNK and phospho-p38 expression levels. This resulted in cell cycle arrest at the G2-M phase and apoptosis in LoVo and RKO cells. Treatment with the JNK inhibitor SP600125 and the p38 MAPK inhibitor SB203580 prevented methyl sartortuoate-induced apoptosis in LoVo cells. Moreover, methyl sartortuoate also prevented neoplasm growth in NOD-SCID nude mice inoculated with LoVo cells. Taken together, these findings suggest that methyl sartortuoate is capable of leading to activation of caspase-8, -9, -3, increasing p53 and Bax/Bcl-2 ratio apoptosis through MAPK-dependent apoptosis and results in G2-M phase arrest in LoVo and RKO cells. Thus, methyl sartortuoate may be a promising anticancer candidate.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Colon/drug effects , Colonic Neoplasms/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Terpenes/therapeutic use , Animals , Anthozoa/chemistry , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Signal Transduction/drug effects , Terpenes/isolation & purification , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Zhonghua Wei Chang Wai Ke Za Zhi ; 17(8): 813-5, 2014 Aug.
Article in Chinese | MEDLINE | ID: mdl-25164901

ABSTRACT

OBJECTIVE: To investigate the safety and feasibility of simultaneous laparoscopic resection of colorectal carcinoma and synchronous liver metastasis. METHODS: Clinical data of 11 patients undergoing simultaneous laparoscopic resection of colorectal carcinoma and synchronous liver metastasis in our hospital from January 2011 to October 2013 were reviewed retrospectively. Feasibility, safety and efficacy of this procedure were investigated. RESULTS: Procedure was completely successful in all the cases without conversion. The mean operation time was (284.6 ± 28.8) min and the mean blood loss was (322.7 ± 75.4) ml. The mean time to intestinal function recovery was (2.9 ± 0.7) d and the mean hospital stay was (12.3 ± 1.9) d. There were no anastomosis leakage, bile leakage, abdominal massive bleeding or infection, and liver failure after operation. During follow-up of 3-35 months, only one patient died of tumor progression. CONCLUSION: Laparoscopic approach for colorectal carcinoma and synchronous liver metastasis is safe and feasible in selected patients.


Subject(s)
Colorectal Neoplasms/surgery , Laparoscopy/methods , Liver Neoplasms/secondary , Adult , Aged , Colectomy/methods , Colorectal Neoplasms/pathology , Female , Hepatectomy , Humans , Liver Neoplasms/surgery , Male , Middle Aged , Retrospective Studies , Treatment Outcome
19.
Oncotarget ; 5(11): 3685-96, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-25003523

ABSTRACT

Our SNP-Chip data demonstrated 7/60 (12%) hepatocellular carcinoma (HCC) patients had PRL-1 copy number amplification. However, its biological functions and signaling pathways in HCC are deficient. Here, we investigated its oncogenic function and prognostic significance in HCC. PRL-1 protein levels were examined in 167 HCC samples by immunohistochemisty (IHC). The relationship of PRL-1 expression and clinicopathological features was assessed by correlation, Kaplan-Meier and Cox regression analyses. The oncogenic function of PRL-1 in HCC cells and its underlying mechanism were investigated by ectopic overexpression and knockdown model. PRL-1 levels in primary HCC and metastatic intravascular cancer thrombus were also determined by IHC. PRL-1 levels were frequently elevated in HCC tissues (81%), and elevated expression of PRL-1 was significantly associated with more aggressive phenotype and poorer prognosis in HCC patients (p<0.05). Ectopic overexpression of PRL-1 markedly enhanced HCC cells migration and invasion. Furthermore, the oncogenic functions of PRL-1 were mediated by PI3K/AKT/GSK3ß signaling pathway through inhibiting E-cadherin expression. Finally, PRL-1 protein levels in metastatic cancer thrombus were higher than that in primary HCC tissues (p<0.05). These data highlight the oncogenic function of PRL-1 in HCC invasion and metastasis implicating PRL-1 as a potential prognostic marker as well as therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Cadherins/metabolism , Cell Movement/physiology , Female , Gene Amplification , Gene Dosage , HEK293 Cells , Hep G2 Cells , Humans , Male , Middle Aged , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
20.
BMC Cancer ; 14: 330, 2014 May 10.
Article in English | MEDLINE | ID: mdl-24885636

ABSTRACT

BACKGROUND: Tumor-associated macrophages (TAMs) are known to promote cancer progression and metastasis through the release of a variety of cytokines. Phosphatase of regenerating liver (PRL-3) has been considered as a marker of colorectal cancer (CRC) liver metastasis. Our previous research suggests that PRL-3 can enhance the metastasis of CRC through the up-regulation of intermediate-conductance Ca2+-activated K+ (KCNN4) channel, which is dependent on the autocrine secretion of tumor necrosis factor-alpha (TNF-α). However, whether TAMs participate in the progression and metastasis of CRC induced by PRL-3 remains unknown. METHODS: We used flow cytometry, coculture, western blotting, invasion assays, real-time quantitative PCR, chromatin immunoprecipitation, luciferase reporter assays, and immunofluorescence staining to determine the effect of TAMs on the ability of PRL-3 to promote invasiveness of CRC cells. RESULTS: In this study, we found that TAMs facilitated the metastasis of CRC induced by PRL-3. When TAMs were cocultured with CRC cells, the expression of KCNN4 was increased in TAMs and the invasion of CRC cells was enhanced. Furthermore, cytokines that were secreted by TAMs, such as IL-6 and IL-8, were also significantly increased. This response was attenuated by treating TAMs with the KCNN4 channel-specific inhibitor, 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34), which suggested that KCNN4 channels may be involved in inducing the secretion of IL-6 and IL-8 by TAMs and improving CRC cell invasiveness. Moreover, the expression of KCNN4 channels in TAMs was regulated through the NF-κB signal pathway, which is activated by TNF-α from CRC cells. Immunofluorescence analysis of colorectal specimens indicated that IL-6 and IL-8 double positive cells in the stroma showed positive staining for the TAM marker CD68, suggesting that TAMs produce IL-6 and IL-8. Increased numbers of these cells correlated with higher clinical stage. CONCLUSIONS: Our findings suggested that TAMs participate in the metastasis of CRC induced by PRL-3 through the TNF-α mediated secretion of IL-6 and IL-8 in a paracrine manner.


Subject(s)
Cell Movement , Colonic Neoplasms/enzymology , Interleukin-6/metabolism , Interleukin-8/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Macrophages/enzymology , Neoplasm Proteins/metabolism , Paracrine Communication , Protein Tyrosine Phosphatases/metabolism , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Macrophages/drug effects , Macrophages/immunology , NF-kappa B/metabolism , Neoplasm Invasiveness , Potassium Channel Blockers/pharmacology , Promoter Regions, Genetic , Signal Transduction , Time Factors , Transfection , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...