Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Cancer ; 12(15): 4585-4594, 2021.
Article in English | MEDLINE | ID: mdl-34149922

ABSTRACT

Purpose: This study aims to develop a liquid biopsy assay to identify HCC and differentially diagnose hepatocellular carcinoma (HCC) from colorectal carcinoma (CRC) liver metastasis. Methods: Thirty-two microRNAs ("HallMark-32" panel) were designed to target the ten cancer hallmarks in HCC. Quantitative PCR and supervised machine learning models were applied to develop an HCC-specific diagnostic model. One hundred thirty-three plasma samples from intermediate-stage HCC patients, colorectal cancer (CRC) patients with liver metastasis, and healthy individuals were examined. Results: Six differentially expressed microRNAs ("Signature-Six" panel) were identified after comparing HCC and healthy individuals. The microRNA miR-221-3p, miR-223-3p, miR-26a-5p, and miR-30c-5p were significantly down-regulated in the plasma of HCC samples, while miR-365a-3p and miR-423-3p were significantly up-regulated. Machine learning models combined with HallMark-32 and Signature-Six panels demonstrated promising performance with an AUC of 0.85-0.96 (p ≤ 0.018) and 0.84-0.93 (p ≤ 0.021), respectively. Further modeling improvement by adjusting sample quality variation in the HallMark-32 panel boosted the accuracy to 95% ± 0.01 and AUC to 0.991 (95% CI 0.96-1, p = 0.001), respectively. Even in alpha fetoprotein (AFP)-negative (< 20ng/mL) HCC samples, HallMark-32 still achieved 100% sensitivity in identifying HCC. The Cancer Genome Atlas (TCGA, n=372) analysis demonstrated a significant association between HallMark-32 and HCC patient survival. Conclusion: To the best of our knowledge, this is the first report to utilize circulating miRNAs and machine learning to differentiate HCC from CRC liver metastasis. In this setting, HallMark-32 and Signature-Six are promising non-invasive tests for HCC differential diagnosis and distinguishing HCC from healthy individuals.

2.
PLoS One ; 12(5): e0177276, 2017.
Article in English | MEDLINE | ID: mdl-28481895

ABSTRACT

PURPOSE: This study aims to establish a highly adaptable workflow downstream of microfluidic enrichment for facilitating systematic CTC enumeration and genetic discovery. METHODS: To facilitate CTC enumeration, we established a CK/EPCAM-combined immunostaining strategy and an automated CTC analytical pipeline using an open-source image analyzer. By virtue of this workflow, we conducted a pilot study of 56 cancer patients and 21 healthy individuals using a high-throughput spiral microfluidic chip system. To facilitate genetic discovery of somatic mutations in CTCs, we integrated the CTC enumeration into next-generation sequencing and established a straightforward amplicon library comprising diversifier random sequences to sequence CTC samples. RESULTS: The CTC staining and enumeration workflow achieved 80.4% sensitivity and 85.7% specificity (AUC = 0.87, p = 0.004, power = 0.985), as evaluated by ROC analysis. Univariate and multivariate analysis verified that the CTC (CK/EpCAM+CD45-), but not other cell populations, is a significant and independent biomarker for cancer patients (p < 0.01). Serial CTC monitoring of the patients revealed reduction in CTC numbers after treatments, suggesting its clinical utility in pharmacodynamic studies. Deep sequencing of CTC samples revealed somatic mutations in TP53 and ESR1. CONCLUSIONS: The significance of this report is to demonstrate a systematic and adaptable workflow to bridge the gap between the microfluidic enrichment and CTC analyses, which fosters broader applications of CTCs in both clinical settings and academic studies.


Subject(s)
Mutation , Neoplastic Cells, Circulating/pathology , Female , Humans , Male , Microfluidic Analytical Techniques
3.
PLoS One ; 7(5): e37159, 2012.
Article in English | MEDLINE | ID: mdl-22606345

ABSTRACT

BACKGROUND: Accumulating evidence supports that tumor growth and cancer relapse are driven by cancer stem cells. Our previous work has demonstrated the existence of CD90(+) liver cancer stem cells (CSCs) in hepatocellular carcinoma (HCC). Nevertheless, the characteristics of these cells are still poorly understood. In this study, we employed a more sensitive RNA-sequencing (RNA-Seq) to compare the gene expression profiling of CD90(+) cells sorted from tumor (CD90(+)CSCs) with parallel non-tumorous liver tissues (CD90(+)NTSCs) and elucidate the roles of putative target genes in hepatocarcinogenesis. METHODOLOGY/PRINCIPAL FINDINGS: CD90(+) cells were sorted respectively from tumor and adjacent non-tumorous human liver tissues using fluorescence-activated cell sorting. The amplified RNAs of CD90(+) cells from 3 HCC patients were subjected to RNA-Seq analysis. A differential gene expression profile was established between CD90(+)CSCs and CD90(+)NTSCs, and validated by quantitative real-time PCR (qRT-PCR) on the same set of amplified RNAs, and further confirmed in an independent cohort of 12 HCC patients. Five hundred genes were differentially expressed (119 up-regulated and 381 down-regulated genes) between CD90(+)CSCs and CD90(+)NTSCs. Gene ontology analysis indicated that the over-expressed genes in CD90(+)CSCs were associated with inflammation, drug resistance and lipid metabolism. Among the differentially expressed genes, glypican-3 (GPC3), a member of glypican family, was markedly elevated in CD90(+)CSCs compared to CD90(+)NTSCs. Immunohistochemistry demonstrated that GPC3 was highly expressed in forty-two human liver tumor tissues but absent in adjacent non-tumorous liver tissues. Flow cytometry indicated that GPC3 was highly expressed in liver CD90(+)CSCs and mature cancer cells in liver cancer cell lines and human liver tumor tissues. Furthermore, GPC3 expression was positively correlated with the number of CD90(+)CSCs in liver tumor tissues. CONCLUSIONS/SIGNIFICANCE: The identified genes, such as GPC3 that are distinctly expressed in liver CD90(+)CSCs, may be promising gene candidates for HCC therapy without inducing damages to normal liver stem cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Adult , Aged , Aged, 80 and over , Base Sequence , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , DNA Primers/genetics , Female , Gene Expression Profiling , Gene Knockdown Techniques , Glypicans/antagonists & inhibitors , Glypicans/genetics , Glypicans/metabolism , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Thy-1 Antigens/metabolism , Tumor Stem Cell Assay
4.
Clin Cancer Res ; 17(10): 3123-33, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21421859

ABSTRACT

PURPOSE: Brain-derived neurotrophic factor (BDNF) has emerged as a novel angiogenic factor, and yet its impact on tumorigenesis is unclear. This study aimed at investigating the roles of BDNF in angiogenesis and tumor development. EXPERIMENTAL DESIGN: BDNF was overexpressed in a mouse endothelial cell (EC) line by stable transfection, and angiogenic properties of the transfectants were assessed. Microarray analysis was employed to explore the molecular pathways. The impact of modulating BDNF levels in two mouse EC lines on tumorigenic potential of a transformed mouse liver cell line was evaluated by an in vivo cotransplantation model. BDNF and tropomyosin receptor kinase B (TrkB) protein levels were determined in 50 pairs of human hepatocellular carcinoma (HCC) tissues by Western blotting and immunohistochemistry. Survival analysis was carried out to determine their clinical significance. RESULTS: Overexpression of BDNF could promote EC proliferation, migration, invasion, and survival. Microarray and molecular studies showed that RhoA, caspase-9, caspase-3, growth arrest specific 6, and VEGF could mediate BDNF/TrkB-induced angiogenesis. The cotransplantation experiment showed that high BDNF-expressing ECs could facilitate tumor angiogenesis and growth, whereas knockdown of BDNF by short hairpin RNAs impaired such effects. Furthermore, examination on human HCC tissues revealed upregulation of BDNF and TrkB protein levels in 46.0% and 33.3% of the cases studied, respectively. Immunohistochemistry disclosed strong BDNF reactivity in both tumor and endothelial cells. High TrkB expression was associated with shorter overall survival. CONCLUSIONS: BDNF/TrkB system was crucial for tumor angiogenesis and growth, which may represent a potential target for antiangiogenic therapy in HCC.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Liver Neoplasms/pathology , Neovascularization, Pathologic/genetics , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , RNA, Small Interfering/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
5.
Protein Pept Lett ; 16(5): 479-85, 2009.
Article in English | MEDLINE | ID: mdl-19442226

ABSTRACT

A monoclonal antibody, McAb9E (IgG3), was generated against a metastatic HCC cell line, MHCC-1. The antigen was characterized as human Caveolin-1 (Cav-1, 21kDa), with pI of 5.65. The Cav-1 antigen was found significantly over expressed in metastatic HCC cell lines as well as in tumor specimens. The Cav-1 specific McAb may be a useful molecular agent for metastatic HCC.


Subject(s)
Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Carcinoma, Hepatocellular/pathology , Caveolin 1/immunology , Proteomics , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Neoplasm Metastasis , Tandem Mass Spectrometry
6.
Cancer Lett ; 273(2): 201-9, 2009 Jan 18.
Article in English | MEDLINE | ID: mdl-18824293

ABSTRACT

The present study investigated the effect of mammalian target of rapamycin (mTOR) inhibition on HCC cells in vitro and in vivo, either alone or in combination with cytotoxic agents. In vitro, HCC cell lines were exposed to RAD001, an mTOR inhibitor, either alone or in combination with cisplatin. Alone, RAD001 suppressed cell proliferation in all cell lines tested, but did not induce apoptosis. RAD001 in combination with cisplatin induced a significant increase in the number of apoptotic cells, downregulated the expression of pro-survival molecules, Bcl-2, survivin and cyclinD1, and increased the cleavage of PARP, compared to RAD001 or cisplatin alone. Transfection of p53 into the Hep3B cell line increased the sensitivity of tumor cells to cisplatin. The suppression of HCC tumor growth in vivo was enhanced by RAD001 combined with cisplatin, accompanied by a significant increase in the number of apoptotic cells in tumor tissues. This study demonstrates that inhibition of mTOR suppresses tumor growth and sensitizes tumor cells to chemocytotoxic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin D1/biosynthesis , Drug Resistance, Neoplasm , Genes, p53 , Humans , Inhibitor of Apoptosis Proteins , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin , TOR Serine-Threonine Kinases
7.
Hepatology ; 47(3): 919-28, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18275073

ABSTRACT

UNLABELLED: Increasing evidence has revealed the importance of cancer stem cells (CSCs) in carcinogenesis. Although liver CSCs have been identified in hepatocellular carcinoma (HCC) cell lines, no data have shown the presence of these cells in human settings. The present study was designed to delineate CSCs serially from HCC cell lines, human liver cancer specimens to blood samples, using CD90 as a potential marker. The number of CD90(+) cells increased with the tumorigenicity of HCC cell lines. CD45(-)CD90(+) cells were detected in all the tumor specimens, but not in the normal, cirrhotic, and parallel nontumorous livers. In addition, CD45(-)CD90(+) cells were detectable in 90% of blood samples from liver cancer patients, but none in normal subjects or patients with cirrhosis. A significant positive correlation between the number of CD45(-)CD90(+) cells in the tumor tissues and the number of CD45(-)CD90(+) cells in the blood samples was identified. CD90(+) cells sorted from cell lines and CD45(-)CD90(+) cells from the tumor tissues and blood samples of liver cancer patients generated tumor nodules in immunodeficient mice. Serial transplantation of CD90(+) cells from tumor xenografts generated tumor nodules in a second and subsequently third batch of immunodeficient mice. Treatment of CD90(+) CSCs with anti-human CD44 antibody induced cell apoptosis in a dose-dependent manner. CONCLUSION: Identification of CD45(-)CD90(+) CSCs in both tumor tissues and circulation suggests that CD45(-)CD90(+) could be used as a marker for human liver cancer and as a target for the diagnosis and therapy of this malignancy.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms/diagnosis , Neoplastic Cells, Circulating/chemistry , Neoplastic Stem Cells/chemistry , Thy-1 Antigens/analysis , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Flow Cytometry , Humans , Leukocyte Common Antigens/analysis , Liver Neoplasms/pathology , Mice , Mice, SCID , Neoplastic Cells, Circulating/pathology , Neoplastic Stem Cells/pathology
8.
Cancer Cell ; 13(2): 153-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242515

ABSTRACT

This study characterized cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) cell lines, tumor specimens, and blood samples. The CD90+ cells, but not the CD90(-) cells, from HCC cell lines displayed tumorigenic capacity. All the tumor specimens and 91.6% of blood samples from liver cancer patients bore the CD45(-)CD90+ population, which could generate tumor nodules in immunodeficient mice. The CD90+CD44+ cells demonstrated a more aggressive phenotype than the CD90+CD44(-) counterpart and formed metastatic lesions in the lung of immunodeficient mice. CD44 blockade prevented the formation of local and metastatic tumor nodules by the CD90+ cells. Differential gene expression profiles were identified in the CD45(-)CD90+ and CD45(-)CD90(-) cells isolated from tissue and blood samples from liver cancer patients and controls.


Subject(s)
Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Thy-1 Antigens/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Separation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Leukocyte Common Antigens/metabolism , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , Neoplasm Transplantation , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
9.
Cancer Biol Ther ; 6(12): 1900-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18059167

ABSTRACT

The present study investigated the effect of YC-1, a novel anti-cancer agent, on the chemo-sensitivity of hepatocellular carcinoma (HCC). YC-1 was administered with chemo-cytotoxic drug, cisplatin, both in vitro and in vivo. YC-1 alone downregulated the expression of phosphorylated form of signal transducers and activators of transcription 3 (P-Stat3[705]), a key mediator in chemo-resistance. When combined with cisplatin, YC-1 further promoted tumor cell apoptosis, decreased the expression of P-Stat3(705), Bcl-xL, CyclinD1 and survivin, and induced the cleavage of caspase 9 and PARP. Overexpression of Stat3 reversed YC-1 induced cell death. YC-1 inhibited Stat3 activity by enhancing the polyubiquitination of P-Stat3(705) induced by cisplatin. In the in vivo setting, YC-1 combined with cisplatin remarkably suppressed tumor growth in a HCC xenograft model, and this effect was also accompanied by YC-1 mediated downregulation of P-Stat3(705), Bcl-xL, Cyclin D1 and survivin, and induction of cleaved caspase 9 and PARP in the tumor tissues. In conclusion, the present study demonstrated a novel anti-cancer effect of YC-1 in enhancing chemo-sensitivity of HCC cells to cisplatin through a Stat3 dependent manner. This finding provides insight into design of a new therapeutic strategy to improve efficacy of chemotherapy in HCC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , Indazoles/pharmacology , Liver Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caspase 9/biosynthesis , Caspase 9/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cyclin D , Cyclins/biosynthesis , Cyclins/genetics , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indazoles/therapeutic use , Inhibitor of Apoptosis Proteins , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational/drug effects , STAT3 Transcription Factor/metabolism , Survivin , Ubiquitination/drug effects , Xenograft Model Antitumor Assays , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
10.
Liver Transpl ; 13(4): 571-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17394163

ABSTRACT

This study aims to investigate the potential role of endogenous interleukin (IL)-10 in long-term liver allograft survival induced by delayed immunosuppression (FK506 days 2-7). Liver transplantation was performed by using Dark Agouti and Lewis rats as donors and recipients, respectively. The delayed immunosuppression protocol induced indefinite allograft survival. A transient upregulation of plasma IL-10 levels was detected in the nontreatment and FK506 treatment groups. Macrophages were found to be one of the major sources of IL-10 produced from the liver allografts. Administration of IL-10-neutralizing antibody shortened the long-term isograft survival and FK506-induced indefinite allograft survival, particularly in the FK506 group. Damaged liver graft histology and increase of plasma alanine aminotransferase levels were detected in the groups with IL-10 antibody treatment. In an ex vivo setting, IL-10 recombinant protein augmented the expression of Foxp3, downregulated the expression of IL-2 and interferon gamma, and induced the generation of CD4(+)CD25(+)Foxp3(+) and CD8(+)CD25(+)Foxp3(+) cells, but this effect was blocked by the administration of IL-10 antibody. Finally, administration of IL-10 recombinant protein after the decline of endogenous IL-10 levels improved allograft survival, and a 100% long-term allograft survival was achieved by the combination of IL-10 with low-dose FK506. In conclusion, the delayed immunosuppression could induce long-term liver allograft survival in the presence of endogenous IL-10 produced by the tissue macrophages. Supplementary exogenous IL-10 administration combined with low-dose immunosuppressive drug may be a useful strategy to induce long-term liver allograft survival.


Subject(s)
Graft Survival/immunology , Immunosuppression Therapy/methods , Interleukin-10/immunology , Liver Transplantation/immunology , Tacrolimus/therapeutic use , Animals , Cell Culture Techniques , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Survival/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Liver Function Tests , RNA/genetics , RNA/isolation & purification , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Tacrolimus/administration & dosage , Time Factors , Transplantation, Homologous
11.
Oncol Rep ; 16(6): 1237-43, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17089044

ABSTRACT

The present study investigated the significance of the serum brain-derived neurotrophic factor (BDNF) and platelets in relation to the clinicopathological features of hepatocellular carcinoma (HCC) patients. Localization of the BDNF expression in human HCC tissues was performed by immunohistochemistry. The measurement of soluble BDNF in the serum was performed by enzyme-linked immunosorbent assay. BDNF was expressed in the cytoplasm of the tumor cells. A positive correlation between the tissue and serum levels of BDNF was identified in the HCC patients. The serum levels of BDNF were positively correlated with the platelet counts in the HCC patients. A higher level of serum BDNF was significantly correlated with a tumor size >5 cm, poorly differentiated HCC, the presence of microsatellite tumor nodules, and the absence of cirrhosis in the non-tumorous tissues. A higher level of the serum BDNF/platelet ratio was associated with a poorer disease-free survival after hepatic resection. This study suggested that the tumor cell was a source of serum BDNF in HCC. A higher serum BDNF level was associated with a more advanced tumor status in the HCC patients. The interaction between serum BDNF and platelets might play an important role in HCC tumor progression.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Platelet Count , Blood Platelets/physiology , Carcinoma, Hepatocellular/mortality , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Liver Neoplasms/mortality , Male , Middle Aged , Prognosis
12.
Mol Cancer Ther ; 5(9): 2261-70, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985060

ABSTRACT

The present study aimed to investigate the therapeutic efficacy of combining vascular endothelial growth factor (VEGF) receptor blockade using tyrosine kinase inhibitor PTK787 with hypoxia for the treatment of hepatocellular carcinoma (HCC). The in vivo effects of the treatments were determined in a rat orthotopic HCC model, in which hypoxia was generated by hepatic artery ligation (HAL). Compared with HAL alone, PTK787 combined with HAL significantly prolonged the animal survival, reduced the tumor size, induced more tumor tissue necrosis and apoptosis, and down-regulated the expression of von Willebrand factor. The mechanism was explored in vitro using murine HCC and endothelial cell lines, respectively. PTK787 combined with hypoxia decreased the expression of VEGF and VEGF receptors in both cell lines and suppressed the cell viability by induction of cell cycle arrest and promotion of apoptosis. Up-regulation of cleaved form caspase-9 and down-regulation of Bcl-2 and cyclin D1 were detected with the combined treatment. Hypoxia sensitized endothelial cells to the inhibitory effect of PTK787 on forming tubular-like structure. The motility of tumor cells was inhibited by hypoxia and the combined approach, with down-regulation of Rac1, Rho, and phosphorylated Akt expression. However, in the endothelial cells, the combined treatment inhibited the hypoxia-enhanced cell motility, with suppressed Rac1, Rho, and phosphorylated Akt expression. In conclusion, PTK787 combined with hypoxia achieved a better therapeutic efficacy than hypoxia alone through enhancing hypoxia-induced antitumor cell effect and preventing the activation of angiogenic process.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Liver Neoplasms, Experimental/drug therapy , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Rats , Rats, Inbred BUF , Vascular Endothelial Growth Factor A/blood
13.
Biochem Biophys Res Commun ; 348(4): 1443-8, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16919599

ABSTRACT

Inhibition of HIF-1alpha activity provides an important strategy for the treatment of cancer. Recently, 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole (YC-1) has been identified as an anti-HIF-1alpha drug in cancer therapy with unclear molecular mechanism. In the present study, we aimed to investigate the effect and mechanism of YC-1 on HIF-1alpha in a hepatocellular carcinoma cell line under hypoxic condition, which was generated by incubating cells with 0.1% O(2). The phenotypic and molecular changes of cells were determined by cell proliferation assay, apoptosis assay, luciferase promoter assay, and Western blot analysis. YC-1 arrested tumor cell growth in a dose-dependent manner, whereas it did not induce cell apoptosis. Hypoxia-induced upregulation of HIF-1alpha was suppressed by YC-1 administration. YC-1 inhibited HIF-1alpha protein synthesis under normoxia and affected protein stability under hypoxia. YC-1 suppressed the expression of total and phosphorylated forms of murine double minute 2 (Mdm2), whereas this inhibitory effect was blocked by overexpression of Mdm2. In conclusion, YC-1 suppressed both protein synthesis and stability of HIF-1alpha in HCC cells, and its inhibitory effects on HIF-1alpha were dependent on Mdm2.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indazoles/pharmacology , Liver Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/physiology , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Cell Line, Tumor , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Liver Neoplasms/pathology
14.
Biochem Biophys Res Commun ; 346(3): 981-5, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16781670

ABSTRACT

Platelets are an important place for the storage of angiogenic factors, such as vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF). The present study aims to investigate the interaction between BDNF-TrkB pathway and platelet activation during tumor development. In an orthotopic hepatocellular carcinoma (HCC) model, increased levels of serum and plasma BDNF were detected with tumor progression. Higher numbers of CD62P+ and TrkB+ platelets were found in the tumor-bearing rats. In the in vitro setting, tumor-conditioned-medium (TCM) and BDNF recombinant protein stimulated CD62P upregulation and subsequent BDNF release in the freshly isolated platelets, whereas this effect could be inhibited by TrkB blockade. TCM and BDNF culture augmented the expression of heat shock protein 90 (Hsp90) in the platelets, which could be reversed by TrkB blockade. In conclusion, this study suggested the presence of BDNF-TrkB autocrine loop in platelets and its importance in regulating platelet activation during tumor development.


Subject(s)
Autocrine Communication , Brain-Derived Neurotrophic Factor/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Platelet Activation , Receptor, trkB/metabolism , Animals , Cell Line, Tumor , Cell Separation , Male , Protein Binding , Rats , Rats, Inbred BUF
15.
Proteomics ; 6(3): 1049-57, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16400691

ABSTRACT

To facilitate the identification of candidate molecular biomarkers that are linked to the pathogenesis of hepatocellular carcinoma (HCC), we investigated protein-expression profiles of 146 tissue specimens including 67 pairs of tumors and adjacent non-tumors resected from HCC patients as well as 12 normal livers by 2-DE. Among the 1800 spots displayed in the liver proteome, a total of 90 protein species were found to be significantly different between the three groups (P < 0.05). Three of the top candidate markers up-regulated in HCC, with high receiver operating characteristic (ROC) curves, were identified by MS/MS analysis and belonged to the chaperone members: heat-shock protein (Hsp)27, Hsp70 and glucose-regulated protein (GRP)78. Over-expression of these chaperone proteins in HCC tissues was confirmed by Western blotting and immunohistochemistry. In correlation with clinico-pathological parameters, expression of Hsp27 was linked to alpha-fetoprotein level (P = 0.007) whereas up-regulation of GRP78 was associated with tumor venous infiltration (P = 0.035). No significant association of Hsp70 with any pathologic features was observed. The present HCC proteome analysis revealed that in response to the stressful cancerous microenvironment, tumor cells strived to increase the expression of chaperone proteins for cyto-protective function and to enhance tumor growth and metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Neoplasm Proteins/metabolism , Proteome/metabolism , Proteomics , Adolescent , Adult , Aged , Blotting, Western , Carcinoma, Hepatocellular/pathology , China , Cohort Studies , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Chaperone BiP , Female , HSP27 Heat-Shock Proteins , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Up-Regulation
16.
Int Immunol ; 17(11): 1391-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16157606

ABSTRACT

Our previous studies revealed that macrophages played an important role in linking injury, inflammatory and immune response in small-for-size liver transplantation. However, the molecular basis that promoted macrophage activation was not clear. In the present study, we explored the potential role of allograft inflammatory factor-1 (AIF-1) in mediating the survival and pro-inflammatory activity of macrophages in a macrophage cell line. First, the expression of AIF-1 was investigated with the stimulation of pro-inflammatory cytokines and anti-inflammatory treatment. Second, the level of inducible nitric oxide synthase (iNOS) and the survival and migration activity of macrophages were determined with the alterations of AIF-1 expression. Finally, a potential molecule that regulated AIF-1 expression was identified by the proteomic approach. The macrophage cell line expressed a certain level of endogenous AIF-1, which could be enhanced by pro-inflammatory cytokines IL-1beta or tumor necrosis factor-alpha and suppressed by anti-inflammatory drug sodium salicylate. AIF-1 augmentation induced by AIF-1/PCDNA3.1(+) transfection enhanced the levels of iNOS and monocyte chemoattractant protein-1, and promoted the cell migration. On the other hand, suppression of AIF-1 expression by AIF-1/short interference RNA (siRNA) inhibited iNOS production, induced macrophage cell apoptosis and blocked the cell migration. Using two-dimensional electrophoresis, a disintegrin and metalloproteinase domain 3 (ADAM3) was identified after AIF-1/siRNA transfection. Transfection of ADAM3/PCDNA3.1(+) up-regulated the expression of AIF-1 and iNOS, whereas suppression of ADAM3 expression down-regulated AIF-1 and iNOS expression. In conclusion, AIF-1 played an important role in the survival and pro-inflammatory activity of macrophages, and ADAM3 might be an upstream molecule that regulated AIF-1 expression.


Subject(s)
Apoptosis/immunology , Calcium-Binding Proteins/immunology , Macrophage Activation/immunology , Macrophages/immunology , Nitric Oxide Synthase Type II/immunology , Animals , Apoptosis/genetics , Calcium-Binding Proteins/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Interleukin-1/immunology , Interleukin-1/pharmacology , Liver Transplantation , Macrophage Activation/genetics , Microfilament Proteins , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Rats , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
17.
Cancer Res ; 65(1): 219-25, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15665298

ABSTRACT

This study aims to identify a novel molecule that may contribute to hepatocarcinogenesis in a rat orthotopic hepatocellular carcinoma model. The hepatocellular carcinoma model was generated by injection of tumor cells into the left lobe of the liver. Proteomic approaches, including ProteinChip and two-dimensional electrophoresis, were used to identify proteins from serially collected rat serum samples. By both ProteinChip and two-dimensional electrophoresis techniques, the level of a 27-kDa protein was found to be augmented in serum samples during tumor development, decreased after left lobectomy, and reincreased at the time of tumor recurrence. The protein was identified to be brain-derived neurotrophic factor (BDNF). By using specific primers and monoclonal antibody, the expression pattern of BDNF was confirmed in tumor tissue but not in the adjacent nontumorous liver tissue. In addition, the truncated isoform of BDNF receptor-tyrosine protein kinase receptor B was only found in tumor tissue. An in vitro study showed that exogenous BDNF could induce tumor cell proliferation predominantly in relatively small numbers of inoculated cells. Administration of BDNF to tumor cell lines induced significantly increased expression of heat shock protein 90 (Hsp90) and cyclin D1, and blocking the activity of Hsp90 could reverse the up-regulation of cyclin D1 induced by BDNF. The present study revealed that BDNF and its receptor were uniquely expressed in tumor tissue and cell lines of hepatocellular carcimona but not in nontumorous liver tissue and normal cell line. BDNF could stimulate tumor cell proliferation in a Hsp90-dependent manner.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms/pathology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Division , Cell Line, Tumor , Cyclin D1/genetics , Disease Models, Animal , HSP90 Heat-Shock Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/genetics , Male , Protein Array Analysis , Rats , Rats, Inbred BUF , Receptor, trkB/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Cancer Lett ; 178(1): 75-81, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11849744

ABSTRACT

Esophageal cancer (ESC) is an important cancer worldwide. Chromosome 11 plays an important role in the development of several human cancers. In this study, 19 polymorphic microsatellite markers spanning the whole chromosome 11 were used to screen for loss of heterozygosity (LOH) in 38 primary esophageal squamous cell carcinomas. Thirty-three of 38 samples (86.8%) showed LOH at one or more loci. High frequencies (51.9-61.1%) of allelic loss were observed at D11S1338, D11S2000, D11S1990, and D11S1647 loci, in two chromosomal regions, 11p15.5 and 11q22.3. The results of this study suggest the presence of putative tumor suppressor genes in these two regions on chromosome 11 related to ESC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 11/genetics , Esophageal Neoplasms/genetics , Loss of Heterozygosity/genetics , Aged , Alcohol Drinking/adverse effects , Female , Genes, Tumor Suppressor , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Staging , Polymerase Chain Reaction , Smoking/adverse effects , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...