Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Exp Cell Res ; 411(1): 112984, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34951997

ABSTRACT

BACKGROUND: Loco-regional invasion is commonly found in oral squamous cell carcinoma (OSCC) and is associated with its poor survival rate. Matrix metalloproteinase-2 (MMP-2) has been implicated in OSCC progression, but its regulation is poorly understood. MATERIALS AND METHODS: Here, one hundred twenty-seven different post-operated human oral cancer tissue samples were analyzed. The messenger RNA (mRNA) expression, protein expression, and MMP-2 activity and MT1-MMP, TIMP-2, and TFs (NFκB, AP1, Sp1, and Twist) were observed semi-quantitative RT-PCR, western blotting, and gelatin zymography. In addition, OSCC derived Cal-27, SCC4/9 cells, photochemical ECGC, and MAPK-pathway inhibitor PD98059 were utilized for in vitro testing and wound healing assay. RESULT: s: Increased protein and activity level of MMP-2 was detected in non-invasive (N0) and invasive (N1-3) oral tumors as compared to the control (adjacent normal) samples. MMP-2 protein and mRNA expression were positively associated with the TFs and MT1-MMP, negatively associated with TIMP-2 expression. Similarly, the MMP-2 expression/activity was related to several signal-transduction pathways like ERK1/2 and wnt-ß-catenin pathways. Treatment of ECGC/MEK inhibitor (PD98059) diminished MMP-2 activity and invasion/migration potential in OSCC. CONCLUSION: Our research suggests that the ERK1/2 driven overexpression/activation of MMP-2 was linked with the overall OSCC invasion and metastasis. Treatment of MEK inhibitor (PD98059) and ECGC diminished MMP-2 activity and thus could be exploited as a therapeutic strategy to control the invasive OSCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Humans , Matrix Metalloproteinase 2/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Prognosis , Survival Rate , Tumor Cells, Cultured
2.
Cell Mol Life Sci ; 79(12): 607, 2022 Nov 27.
Article in English | MEDLINE | ID: mdl-36436127

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide, largely due to the development of colorectal liver metastases (CRLM). For the establishment of CRLM, CRC cells must remodel their tumor-microenvironment (TME), avoid the immune system, invade the underlying stroma, survive the hostile environment of the circulation, extravasate into the liver, reprogram the hepatic microenvironment into a permissive pre-metastatic niche, and finally, awake from a dormant state to grow out into clinically detectable CRLM. These steps form part of the invasion-metastasis cascade that relies on reciprocal interactions between the tumor and its ever-changing microenvironment. Such interplay provides a strong rational for therapeutically targeting the TME. In fact, several TME constituents, such as VEGF, TGF-ß coreceptor endoglin, and CXCR4, are already targeted in clinical trials. It is, however, of utmost importance to fully understand the complex interactions in the invasion-metastasis cascade to identify novel potential therapeutic targets and prevent the establishment of CRLM, which may ultimately greatly improve patient outcome.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Humans , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology , Tumor Microenvironment , Transforming Growth Factor beta , Signal Transduction
3.
Zhonghua Nan Ke Xue ; 29(3): 210-217, 2023 Mar.
Article in Zh | MEDLINE | ID: mdl-38597701

ABSTRACT

OBJECTIVE: To explore the effects of lutein on the adhesion, invasiveness and metastasis of human prostate cancer PC-3M cells and its action mechanism. METHODS: We divided human prostate cancer PC-3M cells into a control, a low-dose lutein, a medium-dose lutein and a high-dose lutein group, and treated them with 0, 10, 20 and 40 µmol/L lutein, respectively. Then we examined the adhesion of the cells to matrix by cell adhesion assay and the changes in cell pseudopodia by Phalloidin staining, detected the expressions of paxillin, matrix metalloproteinase 2 (MMP-2), MMP-9, recombinant tissue inhibitors of metalloproteinase 1 (TIMP-1), E-cadherin, N-cadherin and vimentin by Western blot, determined the invasiveness and migration of the cells by scratch and Transwell assays, and observed their dynamic movement by high-intension imaging. RESULTS: Compared with the control, the lutein intervention groups showed significant reduction in the number of the cells adhered to matrix, the number of cell pseudopodia, the expressions of paxillin, MMP-2, MMP-9, N-cadherin and vimentin, the rates of migration, invasion and metastasis, and the distances of displacement and movement of the cells. However, the expressions of TIMP-1 and epithelial-mesenchymal transition-related E-cadherin were upregulated significantly. CONCLUSION: Lutein can inhibit cell adhesion, reduce the expressions of MMPs, and suppress cell invasion and migration by inhibiting the process of epithelial-mesenchymal transition.


Subject(s)
Matrix Metalloproteinase 2 , Prostatic Neoplasms , Male , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Paxillin/metabolism , Paxillin/pharmacology , Lutein/metabolism , Lutein/pharmacology , Lutein/therapeutic use , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Matrix Metalloproteinase 9/therapeutic use , Vimentin/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Cell Movement , Cell Line, Tumor , Cadherins/metabolism , Cadherins/pharmacology , Cadherins/therapeutic use , Prostatic Neoplasms/pathology , Neoplasm Invasiveness , Epithelial-Mesenchymal Transition
4.
FASEB J ; 34(8): 10623-10639, 2020 08.
Article in English | MEDLINE | ID: mdl-32596959

ABSTRACT

Although renal cell carcinoma (RCC) is a common malignant urological cancer, its pathogenesis remains unclear. Previous studies have indicated that miR-140-5p acts as a tumor suppressor in various tumors, including bladder cancer, hepatocellular carcinoma, and gastric cancer, but its biological function in RCC remains unknown. In the present study, we found that miR-140-5p was upregulated in RCC tissues, whereas Krüppel-like factor 9 (KLF9) was downregulated and correlated inversely with miR-140-5p in RCC tissues. miR-140-5p promoted the proliferation, migration, and invasion of RCC cells in vitro, and knockdown of miR-140-5p significantly suppressed tumor growth and lung metastasis in nude mouse model of RCC. We also found that miR-140-5p significantly suppressed the expression of KLF9 by binding to the 3'-UTR of KLF9 mRNA and that KLF9, as a transcription factor, upregulates KCNQ1 (also called Kv 7.1 and Kv LQT1) expression by binding to the site (-841/-827) in the KCNQ1 promoter region in RCC cells. Moreover, forced expression of KCNQ1 decreased the growth and metastasis of RCC cells. These results suggest that the miR-140-5p/KLF9/KCNQ1 axis functions as a key signaling pathway in RCC progression and metastasis and represents a potential target of RCC therapies.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , KCNQ1 Potassium Channel/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , HEK293 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Up-Regulation/genetics
5.
J Cell Biochem ; 121(1): 231-243, 2020 01.
Article in English | MEDLINE | ID: mdl-31190401

ABSTRACT

The significance of actin-related protein 2/3 complex subunit 4 (ARPC4) expression in bladder cancer, and its potential role in the invasion and migration of bladder cancer cells, has yet to be determined. This study was to identify the correlation between ARPC4 and lymph node metastasis, and to determine the role of ARPC4 in the invasive migration of T24 bladder cancer cells. One hundred and ninety-eight bladder cancer tissues and 40 normal bladder and lymph node tissues were examined. Tissue microarrays were constructed and subjected to immunohistochemical stating for ARPC4. Multiple logistic analysis was used to determine risk factors associated with bladder cancer metastasis. ARPC4 expression in T24 bladder cancer cells was suppressed using small interfering RNA and changes in protein levels were determined by Western blot analysis. The proliferation of bladder cancer cells after knocking down of ARPC4 was determined by cell counting kit-8. The effects of ARPC4 knockdown on T24 cell invasion and migration was determined using transwell and wound healing assays. Immunofluorescence analysis was performed to examine changes in pseudopodia formation and actin cytoskeleton structure. The expression of ARPC4 was elevated in bladder cancer tissues than normal tissues (84.3% vs 27.5%, P < 0.001). The multivariate logistic analysis demonstrated that the level of ARPC4, as a risk factor, was correlated with lymphatic metastasis (P < 0.05). ARPC4 knockdown attenuated proliferation, migration, invasion, and pseudopodia formation in T24 cells. ARPC4 expression, as a risk factor, is associated with lymphatic metastasis and is upregulated in bladder cancer tissues in comparison with normal tissues. Inhibition of ARPC4 expression significantly attenuates the proliferation, migration, and invasion of bladder cancer cell, possibly due to defects in pseudopodia formation.


Subject(s)
Actins/metabolism , Lymphatic Metastasis , Urinary Bladder Neoplasms/metabolism , Actin Cytoskeleton/metabolism , Adult , Aged , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cystectomy , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , RNA, Small Interfering/metabolism , Risk Factors , Tissue Array Analysis , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Wound Healing
6.
BMC Cancer ; 20(1): 626, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32631390

ABSTRACT

BACKGROUND: The failure of treatment for breast cancer usually results from distant metastasis in which the epithelial-mesenchymal transition (EMT) plays a critical role. Hyperinsulinemia, the hallmark of Type 2 diabetes mellitus (T2DM), has been regarded as a key risk factor for the progression of breast cancer. Nuclear receptor subfamily 2, group F, member 2 (NR2F2) has been implicated in the development of breast cancer, however its contribution to insulin-induced EMT in breast cancer remains unclear. METHODS: Overexpression and knockdown of NR2F2 were used in two breast cancer cell lines, MCF-7 and MDA-MB-231 to investigate potential mechanisms by which NR2F2 leads to insulin-mediated EMT. To elucidate the effects of insulin and signaling events following NR2F2 overexpression and knockdown, Cells' invasion and migration capacity and changes of NR2F2, E-cadherin, N-cadherin and vimentin were investigated by real-time RT-PCR and western blot. RESULTS: Insulin stimulation of these cells increased NR2F2 expression levels and promoted cell invasion and migration accompanied by alterations in EMT-related molecular markers. Overexpression of NR2F2 and NR2F2 knockdown demonstrated that NR2F2 expression was positively correlated with cell invasion, migration and the expression of N-cadherin and vimentin. In contrast, NR2F2 had an inverse correlation with E-cadherin expression. In MDA-MB-231, both insulin-induced cell invasion and migration and EMT-related marker alteration were abolished by NR2F2 knockdown. CONCLUSIONS: These results suggest that NR2F2 plays a critical role in insulin-mediated breast cancer cell invasion, migration through its effect on EMT.


Subject(s)
Breast Neoplasms/genetics , COUP Transcription Factor II/metabolism , Epithelial-Mesenchymal Transition/genetics , Insulin/metabolism , Breast Neoplasms/pathology , COUP Transcription Factor II/genetics , Cell Movement/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hyperinsulinism/blood , Hyperinsulinism/epidemiology , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Insulin/blood , MCF-7 Cells , Neoplasm Invasiveness/genetics , Risk Factors
7.
BMC Cancer ; 18(1): 400, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29631554

ABSTRACT

BACKGROUND: The strong invasive and metastatic nature of non-small cell lung cancer (NSCLC) leads to poor prognosis. Collagen triple helix repeat containing 1 (CTHRC1) is involved in cell migration, motility and invasion. The object of this study is to investigate the involvement of CTHRC1 in NSCLC invasion and metastasis. METHODS: A proteomic analysis was performed to identify the different expression proteins between NSCLC and normal tissues. Cell lines stably express CTHRC1, MMP7, MMP9 were established. Invasion and migration were determined by scratch and transwell assays respectively. Clinical correlations of CTHRC1 in a cohort of 230 NSCLC patients were analysed. RESULTS: CTHRC1 is overexpressed in NSCLC as measured by proteomic analysis. Additionally, CTHRC1 increases tumour cell migration and invasion in vitro. Furthermore, CTHRC1 expression is significantly correlated with matrix metalloproteinase (MMP)7 and MMP9 expression in sera and tumour tissues from NSCLC. The invasion ability mediated by CTHRC1 were mainly MMP7- and MMP9-dependent. MMP7 or MMP9 depletion significantly eradicated the pro-invasive effects mediated by CTHRC1 on NSCLC cells. Clinically, patients with high CTHRC1 expression had poor survival. CONCLUSIONS: CTHRC1 serves as a pro-metastatic gene that contributes to NSCLC invasion and metastasis, which are mediated by upregulated MMP7 and MMP9 expression. Targeting CTHRC1 may be beneficial for inhibiting NSCLC metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 9/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Humans , Immunohistochemistry , Lung Neoplasms/mortality , Male , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 9/genetics , NF-kappa B/metabolism , Prognosis , Promoter Regions, Genetic , Proteomics , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction
8.
World J Surg Oncol ; 16(1): 36, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29466986

ABSTRACT

BACKGROUND: KIF18A is associated with a variety of tumours; however, the specific mechanism of action of KIF18A in hepatocellular carcinoma (HCC) remains unclear. In this study, in vitro and in vivo experiments were performed with the aim of exploring the potential function and molecular mechanism of kinesin KIF18A in the occurrence and development of HCC. METHODS: We detected the expression of KIF18A in tumour and adjacent tissues as well as cell proliferation, cell invasion and migration in hepatoma cells after silencing KIF18A. KIF18A-silenced hepatoma cells were subcutaneously injected into nude mice to verify the tumorigenicity of KIF18A. We also detected the expression of signal pathway-related proteins in hepatoma cells after KIF18A knockdown with the aim of exploring the association between KIF18A and related signalling pathways. RESULTS: The level of KIF18A protein was higher in liver cancer tissues than adjacent tissues. After silencing KIF18A in SMMC-7721 and HepG2 cells, cell growth was obviously inhibited; the migration and invasion abilities were significantly decreased and the in vivo tumour weight was decreased compared to the control group (0.201 ± 0.088 g vs 0.476 ± 0.126 g, p = 0.009). The expression of cell cycle-related protein (cyclin B1), invasion and metastasis-related proteins (MMP-7 and MMP-9) and Akt-related proteins in hepatoma cells was also decreased after knocking down KIF18A. CONCLUSIONS: KIF18A may promote proliferation, invasion and metastasis of HCC cells by promoting the cell cycle signalling pathway as well as the Akt and MMP-7/MMP-9-related signalling pathways and may serve as a new target for the diagnosis and treatment of HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/secondary , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Kinesins/metabolism , Liver Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Int J Mol Sci ; 19(4)2018 Apr 08.
Article in English | MEDLINE | ID: mdl-29642503

ABSTRACT

Malignant tumors of the central nervous system (CNS) are among cancers with the poorest prognosis, indicated by their association with tumors of high-level morbidity and mortality. Gliomas, the most common primary CNS tumors that arise from neuroglial stem or progenitor cells, have estimated annual incidence of 6.6 per 100,000 individuals in the USA, and 3.5 per 100,000 individuals in Taiwan. Tumor invasion and metastasis are the major contributors to the deaths in cancer patients. Therapeutic goals including cancer stem cells (CSC), phenotypic shifts, EZH2/AXL/TGF-ß axis activation, miRNAs and exosomes are relevant to GBM metastasis to develop novel targeted therapeutics for GBM and other brain cancers. Herein, we highlight tumor metastasis in our understanding of gliomas, and illustrate novel exosome therapeutic approaches in glioma, thereby paving the way towards innovative therapies in neuro-oncology.


Subject(s)
Cell Movement , Central Nervous System Neoplasms/metabolism , Glioma/metabolism , Animals , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Exosomes/metabolism , Glioma/drug therapy , Glioma/pathology , Humans , MicroRNAs/genetics , Signal Transduction
10.
Breast Cancer Res Treat ; 161(1): 29-40, 2017 01.
Article in English | MEDLINE | ID: mdl-27796714

ABSTRACT

PURPOSE: Cathepsin S (CTSS) is expressed in a variety of cancers and stimulates tumor progression. However, the regulatory mechanism and role of CTSS in breast cancer progression are poorly understood. The aim of this study was to examine the relationships between CTSS expression and breast cancer grade and stage, and the signaling molecules involved in CTSS expression. METHODS: Immunohistochemical staining was performed in tissue microarray sections of 1451 human invasive breast cancer samples to determine epithelial (E-CTSS) and stromal CTSS (S-CTSS) expression. Gene and protein expression levels in human breast cancer cell lines were measured by polymerase chain reaction and western blotting. Small interfering RNA transfection and a Matrigel transwell invasion assay were used to confirm the signaling pathways regulating CTSS expression. RESULTS: In patient tumor tissue blocks, high grade, late stage, and triple negativity were associated with elevated CTSS protein expression, and expression levels were related to the clinical outcomes of patients with invasive breast cancer. CTSS expression was also higher in triple-negative breast cancer (TNBC) cell lines than in hormone-responsive cells, and CTSS expression patterns matched those of tryptophan hydroxylase 1 (TPH1) and 5-hydroxytryptamine receptor 7 (5-HT7). Treatment of TNBC cells (MDA-MB-231 and HCC-1395) with 5-HT significantly enhanced CTSS protein expression, whereas pharmacological inhibition or knockdown of 5-HT7 significantly inhibited its expression. Correspondingly, cancer cell invasion was increased by 5-HT treatment and suppressed by 5-HT7 knockdown. The expression of CTSS was regulated by PI3K/Akt and Ras/Raf/MAPK signaling pathways, and these signaling pathways were stabilized by HSP90 and enhanced by the 5-HT7 receptor-dependent autocrine effect of 5-HT in TNBC cells. CONCLUSION: Our findings suggest CTSS as a candidate target for development of a strategy to inhibit breast cancer invasion, and indicate that HSP90 and 5-HT7 (regulators of CTSS) should be considered as alternative targets for the management of TNBC invasion and metastasis.


Subject(s)
Cathepsins/genetics , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/metabolism , Receptors, Serotonin/metabolism , Serotonin/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Cathepsins/metabolism , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Middle Aged , NF-kappa B/metabolism , Neoplasm Grading , Neoplasm Staging , Phosphatidylinositol 3-Kinases/metabolism , Protein Array Analysis , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/therapy , Tumor Burden , raf Kinases/metabolism , ras Proteins/metabolism
11.
Neoplasma ; 64(3): 389-394, 2017.
Article in English | MEDLINE | ID: mdl-28253718

ABSTRACT

TNF alpha induced protein 3 (TNFAIP3), a member of zinc finger protein family, is a gene whose expression level is promptly induced by the tumor necrosis factor. In this study, the clinical significance of TNFAIP3 was analyzed based on available samples in The Cancer Genome Atlas database. TNFAIP3 downregulation was associated with distant metastasis and worse patient prognosis. TNFAIP3-overexpressing and TNFAIP3-knockdown NPC cell line models were established through plasmid-mediated overexpression and small interfering RNA (siRNA), respectively. Cell migration and invasion capacities were evaluated by wound-healing and transwell assays. Functional studies indicated that TNFAIP3 knockdown promoted migration and invasion, whereas TNFAIP3 overexpression alleviated these functions. Western blot analysis was used to examine protein changes from TNFAIP3 overexpression and knockdown, in which TNFAIP3 promoted the protein expression of E-cadherin and suppressed vimentin expression. Our data suggested that TNFAIP3 inhibited migration and invasion by suppressing epithelial mesenchymal transition in NPC.


Subject(s)
Epithelial-Mesenchymal Transition , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness , RNA, Small Interfering , Vimentin/metabolism
12.
Neoplasma ; 64(1): 101-107, 2017.
Article in English | MEDLINE | ID: mdl-27881010

ABSTRACT

To explore how Tim-3 is expressed and how its expression influences invasion and metastasis of colorectal cancer (CRC) cells. A total of 188 CRC patients were prospectively collected for this study. Meanwhile, 135 normal controls were incorporated during the same period. Intestinal samples of the CRC radical cancerous tissues, paracancerous tissues ( 5.0 cm beyond the cancer tissue) were collected for the following experiment. Furthermore, peripheral venous blood samples (10 ml) were collected from each subject. Immunohistochemical analysis, quantitative real-time polymerase chain reaction (RT-qPCR) and western blot were performed for the detection of Tim-3 in different tissues. The immunohistochemical staining results showed that a positive Tim-3 signal was localized in the cytoplasm and nucleus, observed as yellow or brown granules. Tim-3 was largely expressed in colon carcinoma tissues and normal colon mucosa tissues but was rarely expressed in the cell membrane. RT-qPCR results indicated that Tim-3 mRNA levels were significantly lower in CRC tissues than in paracancerous tissues and normal colon mucosa tissues. A trend of decreased Tim-3 mRNA levels was also found in the paracancerous tissues compared with the normal colon mucosa tissues (all P < 0.05). Western blot results revealed reduced Tim-3 protein expression in CRC tissues compared with normal colon mucosa tissues and paracancerous tissues, and Tim-3 protein expression was much lower in the paracancerous tissues than in the normal colon mucosa tissues (all P < 0.05). Furthermore, obviously lower Tim-3 mRNA levels were found in the poorly differentiated CRC patients and in those with lymph node metastasis and distant metastasis (all P < 0.05). Collectively, Tim-3 expression was mainly located in the cytoplasm and nucleus, showing down-regulated expression in colon carcinoma tissues compared with normal and paracancerous tissues. Reduced Tim-3 expression may promote CRC invasion and metastasis providing a medical reference for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/genetics , Hepatitis A Virus Cellular Receptor 2/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Neoplasm Invasiveness , Neoplasm Metastasis
13.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(4): 566-569, 2017 Jul.
Article in Zh | MEDLINE | ID: mdl-29130678

ABSTRACT

OBJECTIVE: To analyze the expression of lysyl oxidase (LOX) in colorectal cancer and its relationship with clinicopathological characteristics, prognosis, and its progress. METHODS: 82 cases of colorectal tumor paraffin-embedded specimens and paired tumor-adjacent tissues were collected, and data of clinicopathological characteristics and prognosis of these patients were also recorded from 2009.1 to 2010.5. Expressions of LOX, hypoxia inducible factor-1α (HIF-1α), matrix metalloproteinase (MMP)-2, MMP-7 were determined by immunohistochemistry. Then relationship between LOX and clinicopathological characteristics and prognosis was explored, and relationship between LOX and HIF-1α, MMP-2, MMP-7 were investigated. RESULTS: Expressions of LOX was stronger in tumors than in tumor-adjacent tissues (P<0.05). Cancer tissues with overexpressed LOX had later clinical stages, deeper tumor invasion, and more metastatic lymph nodes (all P<0.05). The result also showed that patients with overexpression of LOX had poorer prognosis, and overexpression of LOX was independent factor for prognosis in COX survival analysis. Expression of HIF-1α, MMP-2, MMP-7 in colorectal cancer was stronger than in tumor-adjacent tissues (P<0.05). Positive relationship was found between LOX and HIF-1α, MMP-2, MMP-7 proteins (P<0.05). CONCLUSION: LOX was overexpressed in colorectal cancer tissues, and was associated with the progression of colorectal cancer. LOX may be involved in the progress of colorectal cancer by regulating HIF-1α, MMP-2, MMP-7 protein expression.


Subject(s)
Colorectal Neoplasms/metabolism , Protein-Lysine 6-Oxidase/metabolism , Colorectal Neoplasms/diagnosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 7/metabolism , Prognosis , Survival Analysis
14.
Proteomics ; 15(2-3): 408-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25124533

ABSTRACT

Annexin-1 (ANXA1) is known to be involved in important cellular processes and implicated in cancer. Our previous study showed its roles in cell migration and DNA-damage response processes in breast cancer initiation. In order to understand its roles in tumorigenesis, we extended our studies to analyze tumors derived from polyomavirus middle T-antigen ANXA1 heterozygous (ANXA1(+/-) ) and ANXA1 null (ANXA1(-/-) ) mice. We performed quantitative comparison of ANXA1(+/-) and ANXA1(-/-) tumors employing reductive dimethyl labeling quantitative proteomics. We observed 253 differentially expressed proteins (DEPs) with high statistical significance among over 5000 quantified proteins. Combinatorial use of pathway and network-based computational analyses of the DEPs revealed that ANXA1 primarily modulates processes related to cytoskeletal remodeling and immune responses in these mammary tumors. Of particular note, ANXA1(-/-) tumor showed reduced expression of a known epithelial-to-mesenchymal transition (EMT) marker vimentin, as well as myosin light-chain kinase, which has been reported to induce Rho-kinase mediated assembly of stress fibers known to be implicated in EMT. Integrative network analysis of established interactome of ANXA1 alongside with DEPs further highlights the involvement of ANXA1 in EMT. Functional role of ANXA1 in tumorigenesis was established in invasion assay where knocking down ANXA1 in murine mammary tumor cell line 168FARN showed lower invasive capability. Altogether, this study emphasizes that ANXA1 plays modulating roles contributing to invasion-metastasis in mammary tumorigenesis, distinctive to its roles in cancer initiation.


Subject(s)
Annexin A1/metabolism , Carcinogenesis/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Protein Interaction Maps , Animals , Annexin A1/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mass Spectrometry , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Proteomics
15.
Mol Carcinog ; 53(9): 686-97, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23681900

ABSTRACT

Multidrug resistance (MDR) is one of the most important factors leading to chemotherapeutic failure in patients with breast cancer. The invasive/metastatic ability of MDR cells is strengthened compared with their parental cells. However, the mechanisms underlying MDR have not been fully elucidated. We found that CD44 and the cellular prion protein (PrPc) were both overexpressed in MDR cells (MCF7/Adr and H69AR). Subsequently, we chose the human breast cancer cell line MCF7/Adr, which is resistant to adriamycin, for further research. We discovered that PrPc physically and functionally interacted with CD44. The knockdown of CD44 or PrPc by siRNA in MCF7/Adr cells inhibited cell migration, invasion and proliferation in vitro. However, when the MCF7/Adr cells transfected with CD44 siRNA were incubated with 10 times the peak plasma concentration (PPC) of taxol, their invasive ability was again enhanced. In the breast-carcinoma tissue samples, a significant correlation between the CD44 expression and the PrPc expression was observed in the postneoadjuvant-chemotherapy (NAC) cases. Moreover, in Group 2, which was unresponsive to NAC, the CD44 and PrPc expression levels were significantly increased in the post-NAC cases compared with the pre-NAC cases using the paired-samples t-test. These data indicate that the CD44/PrPc interaction enhances the malignancy of breast cancer cells and affects the responses to neoadjuvant chemotherapy in breast cancer patients. Therefore, blocking the CD44/PrPc interaction may improve outcomes in chemorefractory breast cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Hyaluronan Receptors/metabolism , Neoadjuvant Therapy , PrPC Proteins/metabolism , Adult , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Carcinoma, Medullary/drug therapy , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/genetics , Immunoenzyme Techniques , Middle Aged , Neoplasm Invasiveness , PrPC Proteins/genetics , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
Int J Oncol ; 64(6)2024 06.
Article in English | MEDLINE | ID: mdl-38695241

ABSTRACT

Cancer remains a formidable adversary, challenging medical advancements with its dismal prognosis, low cure rates and high mortality rates. Within this intricate landscape, long non­coding RNAs (lncRNAs) emerge as pivotal players, orchestrating proliferation and migration of cancer cells. Harnessing the potential of lncRNAs as therapeutic targets and prognostic markers holds immense promise. The present comprehensive review delved into the molecular mechanisms underlying the involvement of lncRNAs in the onset and progression of the top five types of cancer. By meticulously examining lncRNAs across diverse types of cancer, it also uncovered their distinctive roles, highlighting their exclusive oncogenic effects or tumor suppressor properties. Notably, certain lncRNAs demonstrate diverse functions across different cancers, confounding the conventional understanding of their roles. Furthermore, the present study identified lncRNAs exhibiting aberrant expression patterns in numerous types of cancer, presenting them as potential indicators for cancer screening and diagnosis. Conversely, a subset of lncRNAs manifests tissue­specific expression, hinting at their specialized nature and untapped significance in diagnosing and treating specific types of cancer. The present comprehensive review not only shed light on the intricate network of lncRNAs but also paved the way for further research and clinical applications. The unraveled molecular mechanisms offer a promising avenue for targeted therapeutics and personalized medicine, combating cancer proliferation, invasion and metastasis.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Neoplasms , RNA, Long Noncoding , RNA, Long Noncoding/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Biomarkers, Tumor/genetics , Cell Proliferation/genetics , Prognosis , Disease Progression
17.
Childs Nerv Syst ; 29(7): 1097-105, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23559392

ABSTRACT

OBJECTS: To determine the mechanism of neuroblastoma (NB) bone invasion/metastasis, it is necessary to investigate the bone invasion/metastasis-related factors in the bone invasion/metastasis process. Some evidence has suggested that various proteins were involved in bone osteolytic response. The invasion/metastasis property and gene expression of NB, however, are still unknown. METHODS: Single-cell suspensions of SY5Y and KCNR cells were injected directly into the femur of nude mice. Radiological and histological analyses, immunohistochemistry analyses, and western blot assay were performed to characterize bone metastasis mechanism in these bone metastasis models. RESULTS: SY5Y and KCNR NB cells result in osteolytic responses in bone metastasis model. Osteoprotegerin (OPG), receptor activator of NF-kappaB ligand (RANKL), parathyroid hormone-related peptide (PTHrP), endothelin 1 (ET-1), and CXCR4 were examined and compared among in vitro, in vivo, and normal bone, respectively. PTHrP, OPG, RANKL, and ET-1 except CXCR4 in SY5Y and KCNR NB cells xenografts were strikingly upregulated compared with normal bone and NB cells. However, significantly stronger expression of PTHrP and RANKL was presented than ET-1 and OPG; furthermore, the ratios of expression of PTHrP, RANKL to OPG, and ET-1 were also markedly increased in vivo versus in vitro. CONCLUSIONS: Our study provided evidence that NB cell may enhance bone invasion through PTHrP, OPG, RANKL, and ET-1, especially PTHrP and RANKL which may display stronger effects. CXCR4 appeared not participating in bone invasion, but in tumor growth, and homing to bone. Targeting PTHrP, OPG, ET-1, and RANKL may provide a new insight and method for patient therapy by inhibiting NB bone metastasis and invasiveness.


Subject(s)
Bone Neoplasms/metabolism , Endothelin-1/metabolism , Neuroblastoma/metabolism , Osteolysis , Osteoprotegerin/metabolism , Parathyroid Hormone-Related Protein/metabolism , RANK Ligand/metabolism , Receptors, CXCR4/metabolism , Animals , Blotting, Western , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary , Cell Line, Tumor , Femur/diagnostic imaging , Femur/pathology , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Neuroblastoma/pathology
18.
Front Oncol ; 13: 1271080, 2023.
Article in English | MEDLINE | ID: mdl-38304037

ABSTRACT

Nasopharyngeal carcinoma (NPC), as one of the most prevalent malignancies in the head and neck region, still lacks a complete understanding of its pathogenesis. Presently, radiotherapy, concurrent chemoradiotherapy, and targeted therapy stand as the primary modalities for treating NPC. With advancements in medicine, the cure rates for nasopharyngeal carcinoma have been steadily increasing. Nevertheless, recurrence and metastasis persist as the primary reasons for treatment failure. Consequently, a profound exploration of the molecular mechanisms underlying the occurrence and progression of nasopharyngeal carcinoma, along with the exploration of corresponding therapeutic approaches, becomes particularly imperative in the quest for comprehensive solutions to combat this disease. High mobility group AT-hook 2 (HMGA2) is a pivotal protein capable of altering chromatin structure, regulating gene expression, and influencing transcriptional activity. In the realm of cancer research, HMGA2 exhibits widespread dysregulation, playing a crucial role in nearly all malignant tumors. It is implicated in various tumorigenic processes, including cell cycle regulation, cell proliferation, epithelial-mesenchymal transition, angiogenesis, tumor invasion, metastasis, and drug resistance. Additionally, HMGA2 serves as a molecular marker and an independent prognostic factor in certain malignancies. Recent studies have increasingly unveiled the critical role of HMGA2 in nasopharyngeal carcinoma (NPC), particularly in promoting malignant progression, correlating with tumor resistance, and serving as an independent adverse prognostic factor. This review focuses on elucidating the oncogenic role of HMGA2 in NPC, suggesting its potential association with chemotherapy resistance in NPC, and proposing its candidacy as an independent factor in nasopharyngeal carcinoma prognosis assessment.

19.
Front Oncol ; 12: 905360, 2022.
Article in English | MEDLINE | ID: mdl-35619916

ABSTRACT

[This corrects the article DOI: 10.3389/fonc.2019.00569.].

20.
Tissue Barriers ; 10(4): 2005420, 2022 10 02.
Article in English | MEDLINE | ID: mdl-34821540

ABSTRACT

E-cadherin is the main component of epithelial adherens junctions (AJs), which play a crucial role in the maintenance of stable cell-cell adhesion and overall tissue integrity. Down-regulation of E-cadherin expression has been found in many carcinomas, and loss of E-cadherin is generally associated with poor prognosis in patients. During the last decade, however, numerous studies have shown that E-cadherin is essential for several aspects of cancer cell biology that contribute to cancer progression, most importantly, active cell migration. In this review, we summarize the available data about the input of E-cadherin in cancer progression, focusing on the latest advances in the research of the various roles E-cadherin-based AJs play in cancer cell dissemination. The review also touches upon the "cadherin switching" in cancer cells where N- or P-cadherin replace or are co-expressed with E-cadherin and its influence on the migratory properties of cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Humans , Epithelial-Mesenchymal Transition/physiology , Cadherins/metabolism , Adherens Junctions/metabolism , Cell Adhesion , Cell Movement , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL