Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
Heliyon ; 9(12): e22586, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38046159

ABSTRACT

SOX8 plays an important role in several physiological processes. Its expression is negatively associated with overall survival in patients with colorectal carcinoma (CRC), suggesting SOX8 is a potential prognostic factor for this disease. However, the role of SOX8 in CRC remains largely unknown. In this study, our data showed that SOX8 expression was upregulated in CRC cell lines and tumor tissues. Stable knockdown of SOX8 in CRC cell lines dramatically reduced cell proliferation, migration, and invasion. Furthermore, the knockdown of SOX8 decreased the phospho-GSK3ß level and suppressed Frizzled-6 (FZD6) transcription; restoration of FZD6 expression partially abolished the effect of SOX8 on Wnt/ß-catenin signaling and promote CRC cell proliferation. In conclusion, our findings suggested that SOX8 served as an oncogene in CRC through the activation of FZD6-dependent Wnt/ß-catenin signaling.

3.
PeerJ ; 11: e16317, 2023.
Article in English | MEDLINE | ID: mdl-38025711

ABSTRACT

Background: Gastric cancer (GC) is an extremely heterogeneous malignancy with a complex tumor microenvironment (TME) that contributes to unsatisfactory prognosis. Methods: The overall activity score for assessing the immune activity of GC patients was developed based on cancer immune cycle activity index in the Tracking Tumor Immunophenotype (TIP). Genes potentially affected by the overall activity score were screened using weighted gene co-expression network analysis (WGCNA). Based on the expression profile data of GC in The Cancer Genome Atlas (TCGA) database, COX analysis was applied to create an immune activity score (IAS). Differences in TME activity in the IAS groups were analyzed. We also evaluated the value of IAS in estimating immunotherapy and chemotherapy response based on immunotherapy cohort. Gene expression in IAS model and cell viability were determined by real-time reverse transcriptase-polymerase chain reaction (RT-qPCR) and Cell Counting Kit-8 (CCK-8) assay, respectively. Results: WGCAN analysis screened 629 overall activity score-related genes, which were mainly associated with T cell response and B cell response. COX analysis identified AKAP5, CTLA4, LRRC8C, AOAH-IT1, NPC2, RGS1 and SLC2A3 as critical genes affecting the prognosis of GC, based on which the IAS was developed. Further RT-qPCR analysis data showed that the expression of AKAP5 and CTLA4 was downregulated, while that of LRRC8C, AOAH-IT1, NPC2, RGS1 and SLC2A3 was significantly elevated in GC cell lines. Inhibition of AKAP5 increased cell viability but siAOAH-IT1 promoted viability of GC cells. IAS demonstrated excellent robustness in predicting immunotherapy outcome and GC prognosis, with low-IAS patients having better prognosis and immunotherapy. In addition, resistance to Erlotinib, Rapamycin, MG-132, Cyclopamine, AZ628, and Sorafenib was reduced in patients with low IAS. Conclusion: IAS was a reliable prognostic indicator. For GC patients, IAS showed excellent robustness in predicting GC prognosis, immune activity status, immunotherapy response, and chemotherapeutic drug resistance. Our study provided novel insights into the prognostic assessment in GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , CTLA-4 Antigen , Prognosis , B-Lymphocytes , Biological Assay , Tumor Microenvironment/genetics , A Kinase Anchor Proteins
4.
J Gastrointest Oncol ; 13(4): 1668-1678, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36092315

ABSTRACT

Background: Previous studies have shown that PD-L1-positive advanced gastric cancer (GC) patients could achieve clinical benefit after receiving immune checkpoint inhibitors (ICI) in initial or subsequent therapy. A number of prospective studies such as Keynote-158 have demonstrated that PD-L1-negative patients who tested as microsatellite instability-high (MSI-H) or tumor mutational burden-high (TMB-H) can benefit from ICIs. In the search for more biomarker for immunotherapy, some studies showed that patients with a specific characteristic to tumor microenvironment (TME) were associated with better prognosis. This study aimed to explore the association between the TME and immunotherapy in PD-L1 negative GC patients. Methods: This study was a retrospective cohort study. Twenty-six CPS PD-L1 negative stage IV advanced GC patients treated with chemoimmunotherapy in Shenzhen Hospital of Peking University were retrospectively enrolled according to the inclusion criteria. Their clinical characteristics were assessed and recorded by independent clinicians. Follow-up data was conducted through the Internet or visit. Respond to treatment was evaluated by RECIST 1.1. The primary outcome was progression-free survival (PFS). The level of tumor-infiltrating lymphocytes (TILs) was measured by multiplex immunofluorescence (mIF) among these patients. Cox proportional hazards analysis was performed to analyzed the correlation between PFS and clinical characteristics including TILs. Results: Among 26 patients, 5 patients (19.2%) were on complete response (CR) and 9 patients (34.6%) were in partial response (PR), while 7 patients (26.9%) experienced stable disease (SD). Intratumoral CD8+ T cells were obviously increased in CPS PD-L1 negative patients who responded to chemoimmunotherapy, compared with patients who did not respond (P=0.011). And higher level of CD8+ TILs was demonstrated to associate with better PFS in CPS PD-L1-negative patients treated with chemoimmunotherapy (HR =23.70, 95% CI: 1.15-488.30, P=0.04). Conclusions: Intratumoral CD8+ TILs may be a potential positive predictive factor of clinical response for chemoimmunotherapy in PD-L1-negative advanced GC. However, the results need to be further confirmed in a cohort with more subjects due to a limited sample sizes in present study.

5.
Crit Rev Eukaryot Gene Expr ; 32(8): 33-42, 2022.
Article in English | MEDLINE | ID: mdl-36017914

ABSTRACT

Colon cancer, as one of the common malignant tumors, has the highest morbidity and mortality. We investigated the clinical significance and possible mechanism of the circular RNA circHIPK2 in the progression of colorectal cancer (CRC). Quantitative analysis of mRNAs, gene microarray hybridization, immunofluorescence, luciferase reporter assay, proliferation assay, EDU staining, subcellular location analysis and Western blotting. circHIPK2 expression was upregulated in patients with CRC compared with paracancerous tissues. In contrast, patients with high circHIPK2 expression had lower overall survival rate and disease-free survival rate than those with low circHIPK2 expression. circHIPK2 expression in normal intestinal epithelial cells was lower than that in CRC cell lines. circHIPK2 promoted CRC progression. miR-485-5p reduced CRC progression. miR-485-5p, as the target of circHIPK2 in CRC model, played a role in promoting CRC progression and expediting HSP90 ubiquitination. HSP90 ubiquitination by miR-485-5p can promote cell proliferation. circHIPK2 has potential clinical significance in CRC progression, which may serve as an exceptional candidate for further therapeutic exploration.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins , Humans , MicroRNAs/metabolism , Ubiquitination
6.
J Cancer Res Clin Oncol ; 148(7): 1721-1735, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34357411

ABSTRACT

PURPOSE: Anlotinib is an anti-angiogenetic multi-targeted tyrosine kinase inhibitor. This study aimed to evaluate the efficacy and safety of anlotinib in advanced non-small cell lung cancer (aNSCLC) in the real world. METHODS: Patients with aNSCLC receiving anlotinib were enrolled in two cohorts (treatment naive and previously treated). The endpoints included progression-free survival (PFS), overall survival (OS) and anlotinib-related adverse events (ar-AEs). RESULTS: 203 patients accrued in the study. In the treatment-naïve cohort (n = 80), the PFS was 7.4 (95% confidence interval [CI] 4.1-10.7) and OS was 10.8 (95% CI 5.8-15.8) months of monotherapy group (immature survival for combination group). In previously treated cohort (n = 123), the PFS was 8.0 months (95% CI 6.1-9.9) in the combination group and 4.3 months (95% CI 2.1-6.6) in the monotherapy group (hazard ratio [HR] 0.49; 95% CI 0.29-0.83; p = 0.007), respectively. The OS was 18.5 months (95% CI 10.5-26.6) in the combination group and 7.8 months (95% CI 7.1-8.4) in the monotherapy group (HR 0.38; 95% CI 0.22-0.66; p = 0.001), respectively. The ar-AEs of grade ≥ 3 in the monotherapy and the combination groups were hypertension (9.0 and 8.7%), fatigue (8.1 and 7.6%), hand-foot syndrome (8.1 and 6.5%), diarrhea (5.4 and 8.7%), proteinuria (5.4 and 5.4%), and mucositis oral (6.3 and 8.7%). CONCLUSION: In aNSCLC, anlotinib monotherapy has a promising efficacy in the first-line setting. It may be an option for those who are ineligible for chemotherapy; anlotinib combination therapy in a ≥ second-line setting showed manageable toxicities and encouraging efficacy, indicating a good application prospect. TRIAL REGISTRATION: This study was retrospectively registered with ISRCTN Registry (ID ISRCTN35543977) on January 26th, 2021 and Chinese Clinical Trial Register (ChiCTR2000032265) on April 4th, 2020.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Quinolines , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Indoles , Quinolines/adverse effects , Treatment Outcome
7.
Transl Cancer Res ; 10(12): 5150-5158, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35116365

ABSTRACT

BACKGROUND: Whether sarcopenia has an impact on immune-related adverse events (irAEs) in patients with malignant neoplasms receiving immune checkpoint inhibitors (ICIs) is not consistent. This study aimed to evaluate the impact of sarcopenia on all grades of irAEs. METHODS: PubMed, Embase, and Cochrane Library databases were systematically searched for related studies up to May 2021. Eligible studies were included according to the PICOS criteria. The risk of bias of the included studies was assessed according to the Newcastle-Ottawa Scale (NOS). The odds ratio (OR), corresponding to the 95% confidence interval (CI) of all grades of irAEs, was collected and analyzed, and a further subgroup analysis of serious adverse events was conducted. All analyses were conducted using the RevMan 5.4 software downloaded from the Cochrane website. The heterogeneity and sensitivity of the study were assessed. RESULTS: Of the 135 references identified, only 8 studies were analyzed, including 519 patients comprising 250 with sarcopenia and 269 without sarcopenia. No obvious bias was observed in the included studies. An increased incidence of irAEs was not observed in patients with sarcopenia at pre-immunotherapy compared to those without sarcopenia. The OR and corresponding 95% CI were 0.97 and 0.62-1.53, respectively (P=0.90), with low heterogeneity (P=0.17, I2 =32%). Further, severe adverse events were analyzed in three studies, and the results showed that sarcopenia was not related to irAEs (P=0.97). CONCLUSIONS: Malignancies with sarcopenia at pre-immunotherapy may not increase the incidence of irAEs, and sarcopenia may not be a predictive factor for irAEs.

8.
Cancer Manag Res ; 12: 6575-6583, 2020.
Article in English | MEDLINE | ID: mdl-32801895

ABSTRACT

PURPOSE: Dexamethasone combined with 5-hydroxytryptamine type 3 receptor antagonists (5-HT3 RA) dual regimen is the standard prophylaxis regimen for patients receiving moderately emetogenic chemotherapy (MEC). However, it has been found in real-world practice that chemotherapy-induced nausea and vomiting (CINV) remains poorly controlled among patients with gastrointestinal tumor, especially in those with high-risk factors for vomiting, such as female, young, and non-alcoholic individuals. Hence, we aimed to evaluate the efficacy of an olanzapine-containing triple regimen in this clinical setting. PATIENTS AND METHODS: We retrospectively reviewed the clinical records of gastrointestinal tumor patients who received mFOLFOX6, XELOX, or FOLFIRI chemotherapy at two institutions. All patients included were female and less than 55 years old, with no history of drinking. The patients were divided into two groups for olanzapine-containing triple therapy (olanzapine, tropisetron, and dexamethasone) and non-olanzapine dual therapy (tropisetron and dexamethasone). The study outcomes were complete response (CR), complete control (CC), nausea control, and quality of life (QoL) by the functional living index-emesis (FLIE) questionnaire. RESULTS: A total of 93 patients were included in the study (olanzapine: 40; control: 53). The CR rate in the olanzapine group was significantly higher than that in the control group in delayed and overall phase (75.0% vs 54.7%, p=0.044; 70.0% vs 47.2%; p=0.028). The CC rate in the overall phase was also better in the olanzapine group (62.5% vs 39.6%, p=0.029). The control of nausea in the overall phase showed a superior trend in the olanzapine group (p=0.059). The olanzapine group exhibited higher FLIE scores, which demonstrated better QoL. More patients in the olanzapine group exhibited somnolence and dizziness. Conversely, the incidence of insomnia and anorexia in the olanzapine group was lower. CONCLUSION: This retrospective study indicates that in gastrointestinal tumor patients with high-risk factors for CINV who were receiving MEC, olanzapine-containing triple antiemetic regimen exhibit better efficacy and QoL as compared to non-olanzapine dual regimen. Further randomized studies are required to confirm these results.

9.
J Cell Physiol ; 235(4): 3579-3591, 2020 04.
Article in English | MEDLINE | ID: mdl-31552684

ABSTRACT

HOXC10 plays a critical role in many cellular processes, such as proliferation, migration, and invasion, but the function of HOXC10 in gastric carcinoma is not clear. In this study, we aimed to investigate the expression profile of HOXC10 and its role in gastric carcinoma cells and in vivo experiments. HOXC10 expression patterns were detected in clinical samples and gastric cancer cells lines by reverse transcriptase polymerase chain reaction assays, and then, we focused on its role in regulating cell proliferation, cell cycle, migration, and invasion after transfection of silencing and overexpression plasmids in vitro and in vivo. Finally, we confirmed the correlation between HOXC10 and nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), transforming growth factor-ß (TGF-ß), and epidermal growth factor receptor expression. We found that HOXC10 expression increased in clinical samples, especially in poorly differentiated (PD) gastric cancer cells. Silencing HOXC10 suppressed proliferation, migration, and invasion in vitro, and inhibited tumor growth and induced apoptosis in vivo. Overexpression of HOXC10 showed the opposite effect on PD gastric cancer cells. In addition, silencing HOXC10 inhibited the expression of interleukin-6, TNF-α, TGF-ß, and epidermal growth factor, and overexpressing HOXC10 induced their expression both in vitro and in vivo. Luciferase reporter assays and chromatin immunoprecipitation indicated that HOXC10 may activate the NF-κB signaling pathway through regulation of P65 transcriptional activity by binding to the P65 promoter. HOXC10 may play an important role in PD gastric carcinoma cell proliferation, cell cycle, migration, invasion, and metastasis through upregulating proinflammatory cytokines via NF-κB pathway, suggesting HOXC10 may serve as a novel therapeutic target for PD gastric cancer.


Subject(s)
Carcinoma/genetics , Cell Proliferation/genetics , Homeodomain Proteins/genetics , Stomach Neoplasms/genetics , Animals , Carcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Cytokines/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Interleukin-6/genetics , Lymphotoxin-alpha/genetics , Male , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Stomach Neoplasms/pathology , Tumor Necrosis Factor-alpha/genetics
10.
Onco Targets Ther ; 12: 7887-7896, 2019.
Article in English | MEDLINE | ID: mdl-31576146

ABSTRACT

BACKGROUND: Advanced gastric cancer (aGC) has a high global incidence and a high mortality rate and because of its high malignancy and heterogeneity, the existing methods for prognosis are limited, and a new treatment model is necessary. Circulating tumor cells (CTCs) could be considered as a "liquid biopsy" for tumor diagnosis and for monitoring treatment responses and predicting clinical outcome. Clinical studies support the efficacy of programmed cell death 1 (PD-1) immunotherapy in a subset of aGC. METHODS: Cell capture efficiency, as described by the CanPatrol CTC enrichment technique, was validated using artificial blood samples as well as blood samples from 32 aGC patients. Clinicopathologic data of patients were collected from the hospital information system. We used CanPatrol for CTC isolation, classification, and clinical analysis. RESULTS: A cell capture efficiency of >80% was achieved. Significant correlation was observed between CTC enumeration and clinicopathology parameters, including the Lauren classification (r=0.470, P=0.008), perineural invasion (r=0.393, P=0.029), TNM stage (r=0.740, P<0.001), and Ki-67 level (r=0.510, P=0.005). When compared to the traditional methods, monitoring CTC subtypes exhibits higher sensitivity of evaluating the disease status. Enumeration of epithelial CTC subset and its relative abundance in the total CTC pool are highly correlated with clinical efficacy. CTC programmed cell death ligand-1 (PD-L1) could be successfully detected for immunotherapy in addition to PD-L1 immunohistochemistry and microsatellite instability. CONCLUSION: We provide a new method that allows for the simple and effective detection of CTCs in aGC. It has potential clinical applications in monitoring prognosis and guiding future individualized immunotherapy.

11.
Cancer Med ; 8(16): 7044-7054, 2019 11.
Article in English | MEDLINE | ID: mdl-31557409

ABSTRACT

BACKGROUND: Immunotherapy and its mechanisms are being studied in a wide variety of cancers. Programmed cell death ligand 1 (PDL1) is associated with immune evasion in numerous tumor types. Here, we aimed to assess the relationship between metastasis associated in colon cancer-1 (MACC1) and PDL1 and examine their effects on gastric cancer (GC) tumor immunity. METHODS: The expression of MACC1, c-Met, and PDL1 in human GC tissues was first assessed using quantitative RT-PCR (qRT-PCR) and immunohistochemistry. We then focused on the relationships among MACC1, c-Met, and PDL1 using RT-PCR and western blotting after cell transfection and inhibitor treatment in vitro and on the identification of their roles in immune killing in vitro and in vivo. RESULTS: We found that expression of MACC1, c-Met, and PDL1 was upregulated in human GC tissues, and there was a positive correlation between the expression levels. In addition, we found that ectopic expression of MACC1 (silencing and overexpression by transfection) resulted in corresponding changes in c-Met and PDL1 expression levels, and c-Met/AKT/mTOR pathway inhibitors (SU11274, MK2206, and rapamycin) blocked the regulation of PDL1 expression by MACC1. Furthermore, silencing of MACC1 led to an increase in antitumor and immune killing in vitro and in vivo, and overexpression of MACC1 resulted in a decrease in tumor immunity in vitro and in vivo. CONCLUSIONS: From these data, we infer that MACC1 regulates PDL1 expression and tumor immunity through the c-Met/AKT/mTOR pathway in GC cells and suggest that MACC1 may be a therapeutic target for GC immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Animals , B7-H1 Antigen/genetics , Cell Line, Tumor , Cytokines/immunology , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Mice, Nude , Middle Aged , Proto-Oncogene Proteins c-met/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Trans-Activators/genetics
12.
Surg Endosc ; 33(11): 3550-3557, 2019 11.
Article in English | MEDLINE | ID: mdl-31342257

ABSTRACT

BACKGROUND: The purpose of this study is to compare the clinical outcomes of laparoscopic liver resection versus open liver resection for recurrent hepatocellular carcinoma (RHCC). METHODS: Published studies which investigated laparoscopic versus open liver resection for RHCC were identified, and meta-analysis was used for statistical analysis. RESULTS: Six studies were analyzed by meta-analysis method, and cumulative 335 cases were included in this study. Laparoscopic liver resection was performed in 145 cases, and open liver resection was performed in 190 cases. Meta-analysis showed that there was no difference in operative time and 90-day mortality between the laparoscopic group and the open group (p = 0.06 and p = 0.06 respectively); Nevertheless, compared with the open group, the laparoscopic group resulted in significantly lower rate of in-hospital complication (p < 0.0001), much less blood loss (p < 0.0001) and shorter postoperative hospital stay (p = 0.002). CONCLUSION: Laparoscopic liver resection for RHCC offers a benefit of lower in-hospital complication rate, less blood loss, shorter postoperative hospital stay, while similar operative time and 90-day mortality as the open liver resection. Laparoscopic liver resection is feasible with satisfactory postoperative outcomes and can be a safe alternative treatment strategy to open procedure for RHCC.


Subject(s)
Carcinoma, Hepatocellular/surgery , Liver Neoplasms/surgery , Neoplasm Recurrence, Local/surgery , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Hepatectomy/methods , Humans , Laparoscopy/methods , Length of Stay , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Open Abdomen Techniques/methods , Operative Time , Postoperative Complications , Retrospective Studies , Treatment Outcome
13.
IEEE Trans Biomed Eng ; 66(6): 1536-1541, 2019 06.
Article in English | MEDLINE | ID: mdl-30307854

ABSTRACT

OBJECTIVE: We have developed a novel simple wedge-shaped microfluidic device for highly efficient isolation of circulating tumor cells (CTCs) from cancer patient blood samples. METHODS: We used wet chemical etching and thermal bonding technologies to fabricate the wedge-shaped microdevice and performed optimization assays to obtain optimal capture parameters. Cancer cells spiked samples were used to evaluate the capture performance. Clinical assays were performed to isolate and identify CTCs from whole blood samples of patients with liver, breast, lung, and gastric cancer. RESULTS: Outlet height of 5.5 µm and flow rate of 200 µL/min were chosen as the optimal CTC-capture conditions. This method exhibited excellent isolation performance (more than 85% capture efficiency) for four cancer cell lines (HepG2, SKBR3, A549, and BGC823). In clinical assay, the platform identified CTCs 5 in 6 liver (83.3%), 8 in 10 breast (80%), 5 in 8 lung (62.5%), 5 in 9 gastric (55.6%) cancer patients, and only 1 in 25 healthy blood samples (4%). CONCLUSION: Our wedge-shaped microfluidic device had several advantages, including relatively simple fabrication, high capture efficiency, simple sample processing steps, and easy observation. SIGNIFICANCE: This method had successfully demonstrated the clinical feasibility of CTC isolation and shown a great potential of clinical usefulness in monitoring tumor prognosis and guiding individualized treatment in the future.


Subject(s)
Cell Separation/instrumentation , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Neoplastic Cells, Circulating , Cell Line, Tumor , Cell Separation/methods , Equipment Design , Humans , Microfluidic Analytical Techniques/methods , Neoplasms/diagnosis , Neoplasms/pathology
14.
Oncol Lett ; 15(3): 3971-3976, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29467907

ABSTRACT

Type 1 transforming growth factor ß receptor (TGFBR1)*6A, a common hypomorphic variant of TGFBR1, may act as a susceptibility allele in colorectal cancer. However, the contribution of TGFBR1*6A to colorectal cancer development is largely unknown. To test the hypothesis that TGFBR1*6A promotes colorectal cancer invasion and metastasis via Smad-independent transforming growth factor-ß (TGF-ß) signaling, the effect of TGFBR1*6A on the invasion of colorectal cancer cells was assessed. pCMV5-TGFBR1*6A-HA plasmids were transfected into SW48 and DLD-1 cells by Lipofectamine-mediated DNA transfection. The effect of TGF-ß1 on the proliferation of SW48 and DLD-1 cells transfected with TGFBR1*6A was determined by MTT assay. The effects of the TGF-ß1 on the invasion of the transfected SW48 and DLD-1 cells were determined using Matrigel-coated plates. Transforming migrating chambers were used to determine the effects of TGF-ß1 on the migration of the transfected SW48 and DLD-1 cells. Western blot analysis was used to determine the expression of phosphorylated (p-) extracellular-signal-regulated kinase (ERK), p-P38 and p-SMAD family member 2 in SW48 cells. Using transfected TGFBR1*6A SW48 and DLD-1 cell lines our group demonstrated that, in comparison with TGFBR1*9A, TGFBR1*6A is capable of switching TGF-ß1 growth-inhibitory signals into growth-stimulatory signals which significantly increased the invasion of SW48 and DLD-1 cells. Functional assays indicated that TGFBR1*6A weakened Smad-signaling but increased ERK and p38 signaling, which are crucial mediators of cell migration and invasion. From this, it was possible to conclude that TGFBR1*6A enhanced SW48 cell migration and invasion through the mitogen-activated protein kinase pathway and that it may contribute to colorectal cancer progression in a TGF-ß1/Smad signaling-independent manner. This suggests that TGFBR1*6A may possess oncogenic properties and that it may affect the migration and invasion of colorectal cancer cells.

15.
Am J Transl Res ; 9(10): 4545-4552, 2017.
Article in English | MEDLINE | ID: mdl-29118916

ABSTRACT

Colorectal cancer (CRC) remains one of the most common cancers worldwide. Increasing evidence indicates that long non-coding RNAs (lncRNAs) regulate diverse cellular processes, including cell growth, differentiation, apoptosis, and cancer progression. However, the function of lncRNAs in the progression of CRC remains largely unknown. Here, we reported that HOXA cluster antisense RNA2 (HOXA-AS2) was upregulated in CRC. Increased HOXA-AS2 expression in CRC was associated with larger tumor size and higher clinical stage. In vitro experiments revealed that HOXA-AS2 knockdown significantly inhibited CRC cell proliferation by causing G1 arrest and promoting apoptosis, whereas HOXA-AS2 overexpression promoted cell proliferation. Further functional assays indicated that HOXA-AS2 overexpression significantly promoted cell migration and invasion by regulating the epithelial-mesenchymal transition (EMT). In conclusion, our study identifies HOXA-AS2C as a potential biomarker in CRC.

16.
Biomed Microdevices ; 19(4): 93, 2017 Oct 25.
Article in English | MEDLINE | ID: mdl-29071494

ABSTRACT

Circulating tumor cells (CTCs) have been regarded as the major cause of metastasis, holding significant insights for tumor diagnosis and treatment. Although many efforts have been made to develop methods for CTC isolation and release in microfluidic system, it remains significant challenges to realize highly efficient isolation and gentle release of CTCs for further cellular and bio-molecular analyses. In this study, we demonstrate a novel method for CTC isolation and release using a simple wedge-shaped microfluidic chip embedding degradable znic oxide nanorods (ZnNRs) substrate. By integrating size-dependent filtration with degradable nanostructured substrate, the capture efficiencies over 87.5% were achieved for SKBR3, PC3, HepG2 and A549 cancer cells spiked in healthy blood sample with the flow rate of 100 µL min-1. By dissolving ZnNRs substrate with an extremely low concentration of phosphoric acid (12.5 mM), up to 85.6% of the captured SKBR3 cells were released after reverse injection with flow rate of 100 µL min-1 for 15 min, which exhibited around 73.6% cell viability within 1 h after release to around 93.9% after re-cultured for 3 days. It is conceivable that our microfluidic device has great potentials in carrying on cell-based biomedical studies and guiding individualized treatment in the future.


Subject(s)
Lab-On-A-Chip Devices , Nanotubes/chemistry , Neoplastic Cells, Circulating/chemistry , Zinc Oxide/chemistry , A549 Cells , Cell Line, Tumor , Cell Survival/drug effects , Hep G2 Cells , Humans , Microfluidic Analytical Techniques
17.
Oncotarget ; 8(2): 3029-3041, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27935872

ABSTRACT

INTRODUCTION: Circulating tumor cells (CTCs) play a crucial role in cancer metastasis. In this study, we introduced a novel isolation method by size of epithelial tumor cells (ISET) device with automatic isolation and staining procedure, named one-stop ISET (osISET) and validated its feasibility to capture CTCs from cancer patients. Moreover, we aim to investigate the correlation between clinicopathologic features and CTCs in colorectal cancer (CRC) in order to explore its clinical application. RESULTS: The capture efficiency ranged from 80.3% to 88% with tumor cells spiked into medium while 67% to 78.3% with tumor cells spiked into healthy donors' blood. In detection blood samples of 72 CRC patients, CTCs and clusters of circulating tumor cells (CTC-clusters) were detected with a positive rate of 52.8% (38/72) and 18.1% (13/72) respectively. Moreover, CTC positive rate was associated with factors of lymphatic or venous invasion, tumor depth, lymph node metastasis and TNM stage in CRC patients (p < 0.01). Lymphocyte count and neutrophil to lymphocyte ratio (NLR) were significantly different between CTC positive and negative groups (p < 0.01). MATERIALS AND METHODS: The capture efficiency of the device was tested by spiking cancer cells (MCF-7, A549, SW480, Hela) into medium or blood samples of healthy donors. Blood samples of 72 CRC patients were detected by osISET device. The clinicopathologic characteristics of 72 CRC patients were collected and the association with CTC positive rate or CTC count were analyzed. CONCLUSIONS: Our osISET device was feasible to capture and identify CTCs and CTC-clusters from cancer patients. In addition, our device holds a potential for application in cancer management.


Subject(s)
Early Detection of Cancer/instrumentation , Early Detection of Cancer/methods , Neoplasms/diagnosis , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Count , Cell Line, Tumor , Cell Separation/instrumentation , Cell Separation/methods , Female , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Sensitivity and Specificity , Staining and Labeling/instrumentation , Staining and Labeling/methods
18.
Theranostics ; 6(11): 1877-86, 2016.
Article in English | MEDLINE | ID: mdl-27570557

ABSTRACT

Circulating tumor cells (CTCs) have been considered as the origin of cancer metastasis. Thus, detection of CTCs in peripheral blood is of great value in different types of solid tumors. However, owing to extremely low abundance of CTCs, detection of them has been technically challenging. To establish a simple and efficient method for CTCs detection in patients with hepatocellular carcinoma (HCC), we applied biocompatible and transparent HA/CTS (Hydroxyapatite/chitosan) nanofilm to achieve enhanced topographic interactions with nanoscale cellular surface components, and we used sLex-AP (aptamer for carbohydrate sialyl Lewis X) to coat onto HA/CTS nanofilm for efficient capture of HCC CTCs, these two functional components combined to form our CTC-(BioT)Chip platform. Using this platform, we realized HCC CTCs' capture and identification, the average recovery rate was 61.6% or more at each spiking level. Importantly, our platform identified CTCs (2±2 per 2 mL) in 25 of 42 (59.5%) HCC patients. Moreover, both the positivity rate and the number of detected CTCs were significantly correlated with tumor size, portal vein tumor thrombus, and the TNM (tumor-node-metastasis) stage. In summary, our CTC-(BioT)Chip platform provides a new method allowing for simple but efficient detection of CTCs in HCC patients, and it holds potential of clinically usefulness in monitoring HCC prognosis and guiding individualized treatment in the future.


Subject(s)
Aptamers, Nucleotide/chemistry , Carcinoma, Hepatocellular/diagnosis , Cell Separation/methods , Liver Neoplasms/diagnosis , Nanostructures/chemistry , Neoplastic Cells, Circulating , Surface Properties , Carcinoma, Hepatocellular/secondary , Cell Adhesion , Chitosan , Durapatite , Humans , Liver Neoplasms/secondary
19.
J Cancer ; 7(1): 69-79, 2016.
Article in English | MEDLINE | ID: mdl-26722362

ABSTRACT

BACKGROUND: Capture and identification of circulating tumor cells (CTCs) in the blood system can help guide therapy and predict the prognosis of cancer patients. However, simultaneous capture and identification of CTCs with both epithelial and mesenchymal phenotypes remains a formidable technical challenge for cancer research. This study aimed at developing a system to efficiently capture and identify these CTCs with heterogeneous phenotypes using transparent nanomaterials and quantum dots (QDs)-based multiplexed imaging. METHODS: Hydroxyapatite-chitosan (HA-CTS) nanofilm-coated substrates were modified based on our previous work to increase the capture efficiency of cancer cell lines by extending baking and incubating time. QDs-based imaging was applied to detect cytokeratin, epithelial cell adhesion molecule (EpCAM), and vimentin of cancer cells to demonstrate the feasibility of multiplexed imaging. And QDs-based multiplexed imaging of CD45, cytokeratin and vimentin was applied to detect CTCs from different cancer patients that were captured using HA-CTS nanofilm substrates. RESULTS: Comparisons of the capture efficiencies of cancer cells at different baking time of film formation and incubating time of cell capture revealed the optimal baking and incubating time. Optimal time was chosen to develop a modified CTCs capture system that could capture EpCAM-positive cancer cells at an efficiency > 80%, and EpCAM-negative cancer cells at an efficiency > 50%. QDs-based imaging exhibited comparable detection ability but higher photostability compared to organic dyes imaging in staining cells. In addition, QDs-based multiplexed imaging also showed the molecular profiles of cancer cell lines with different phenotypes well. The integrated CTCs capture and identification system successfully captured and imaged CTCs with different sub-phenotypes in blood samples from cancer patients. CONCLUSION: This study demonstrated a reliable capture and detection system for heterogeneous CTCs that combined enrichment equipment based on HA-CTS nanofilm substrates with QDs-based multiplexed imaging.

20.
Technol Cancer Res Treat ; 15(1): 69-76, 2016 Feb.
Article in English | MEDLINE | ID: mdl-24988051

ABSTRACT

Cancer cells that detach from solid tumor and circulate in the peripheral blood (CTCs) have been considered as a new "biomarker" for the detection and characterization of cancers. However, isolating and detecting cancer cells from the cancer patient peripheral blood have been technically challenging, owing to the small sub-population of CTCs (a few to hundreds per milliliter). Here we demonstrate a simple and efficient cancer cells isolation and purification method. A biocompatible and surface roughness controllable TiO2 nanofilm was deposited onto a glass slide to achieve enhanced topographic interactions with nanoscale cellular surface components, again, anti-CD45 (a leukocyte common antigen) and anti-EpCAM (epithelial cell adhesion molecule) were then coated onto the surface of the nanofilm for advance depletion of white blood cells (WBCs) and specific isolation of CTCs, respectively. Comparing to the conventional positive enrichment technology, this method exhibited excellent biocompatibility and equally high capture efficiency. Moreover, the maximum number of background cells (WBCs) was removed, and viable and functional cancer cells were isolated with high purity. Utilizing the horizontally packed TiO2 nanofilm improved pure CTC-capture through combining cell-capture-agent and cancer cell-preferred nanoscale topography, which represented a new method capable of obtaining biologically functional CTCs for subsequent molecular analysis.


Subject(s)
Cell Separation/methods , Cell Adhesion , HCT116 Cells , HeLa Cells , Humans , Immobilized Proteins/chemistry , Leukocyte Common Antigens/chemistry , Metal Nanoparticles/chemistry , Neoplastic Cells, Circulating , Titanium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...