Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancer Med ; 13(4): e7018, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38457189

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Hepatectomy remains the first-line treatment for patients with resectable HCC. However, the reported recurrence rate of HCC at 5 years after surgery is between 50% and 70%. Tumor-related factors, including tumor size, number and differentiation, and underlying liver disease are well-known risk factors for recurrence after treatment. In addition to tumor-related factors, ever-increasing amounts of studies are finding that the tumor microenvironment also plays an important role in the recurrence of HCC, including systemic inflammatory response and immune regulation. Based on this, some inflammatory and immune markers were used in predicting postoperative cancer recurrence. These include neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, cytotoxic T cells, and regulatory T cells, among others. In this review, we summarized the inflammatory and immune markers that affect recurrence after HCC resection in order to provide direction for adjuvant therapy after HCC resection and ultimately achieve the goal of reducing recurrence.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Hepatectomy/adverse effects , Liver Neoplasms/pathology , Neoplasm Recurrence, Local/surgery , Inflammation/etiology , Lymphocytes/pathology , Biomarkers , Retrospective Studies , Prognosis , Tumor Microenvironment
2.
ACS Appl Mater Interfaces ; 16(7): 8391-8402, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38324389

ABSTRACT

Enriching erythrocytes and platelets in seconds and providing a fast seal in bleeding sites is vital to fatal hemorrhage control. Herein, hydrophilic chitosan fibrous mats (CECS-D mats) are fabricated by introducing hydrophilic carboxyethyl groups and subsequent catechol groups onto chitosan fibers. Due to strong hydrophilicity, CECS-D mats exhibit rapid liquid-absorption capacity, especially instantaneous absorptivity to the rabbit blood, which can achieve erythrocyte and platelet aggregations quickly by concentrating blood, thus promoting the formation of blood clots. Furthermore, the mats are self-oxidated to form quinone-amine adducts or quinone multimers by adjusting pH conditions, which not only provides tissue adhesion but also induces erythrocyte aggregation and platelet adhesion, further enhancing the seal and triggering quick closure to achieve fast hemostasis. Therefore, the mats reveal superior hemostatic performance in rabbit liver and spleen models over CECS mats and gauze. Especially in the fatal femoral artery injury model of rabbits, the mats reduce the blood loss by ∼75% and shortened the bleeding time by ∼50% compared with CECS mats, which have been reported to have the same hemostatic effect as commercialized Celox products in a swine femoral artery injury model. Besides, the mats are cytocompatible and degradable as well as antibacterial. This chitosan mat is a promising hemostatic material for fatal hemorrhage control.


Subject(s)
Chitosan , Hemostatics , Rabbits , Animals , Swine , Chitosan/pharmacology , Hemorrhage/drug therapy , Hemostatics/pharmacology , Hemostasis , Hydrophobic and Hydrophilic Interactions , Quinones
3.
Mol Immunol ; 165: 42-54, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38150981

ABSTRACT

OBJECTIVE: Cells can produce stress granules (SGs) to protect itself from damage under stress. The cGAS-STING pathway is one of the important pattern recognition pathways in the natural immune system. This study was investigated whether human mesenchymal stem cells (hMSCs) could protect the liver by inducing M2 macrophages to produce SGs during acute drug induced liver injury (DILI) induced by acetaminophen (APAP). METHODS: After intragastric administration of APAP in vivo to induce DILI mice model, hMSCs were injected into the tail vein. The co-culture system of hMSCs and M2 macrophages was established in vitro. It was further use SGs inhibitor anisomicin to intervene M2 macrophages. The liver histopathology, liver function, reactive oxygen species (ROS) level, apoptosis pathway, endoplasmic reticulum stress (ERS) level, SGs markers (G3BP1/TIA-1), cGAS-STING pathway, TNF-α, IL-6, IL-1ß mRNA levels in liver tissue and M2 macrophages were observed. RESULTS: In vivo experiments, it showed that hMSCs could alleviate liver injury, inhibite the level of ROS, apoptosis and ERS, protect liver function in DILI mice. The mount of M2 was increased in the liver. hMSCs could also induce the production of SGs, inhibit the cGAS-STING pathway and reduce TNF-α, IL-6, IL-1ß mRNA expression. The results in vitro showed that hMSCs could induce the production of SGs in macrophages, inhibit the cGAS-STING pathway, promote the secretion of IL-4 and IL-13, and reduce TNF-α, IL-6, IL-1ß mRNA level in cells. In the process of IL-4 inducing M2 macrophage activation, anisomycin could inhibit the production of SGs, activate the cGAS-STING pathway, and promote the inflammatory factor TNF-α, IL-6, IL-1ß mRNA expression in cells. CONCLUSIONS: HMSCs had a protective effect on acute DILI in mice induced by APAP. Its mechanism might involve in activating M2 type macrophages, promoting the production of SGs, inhibiting the cGAS-STING pathway, and reducing the expression of pro-inflammatory factors in macrophages, to reduce hepatocytes damage.


Subject(s)
Chemical and Drug Induced Liver Injury , Mesenchymal Stem Cells , Humans , Mice , Animals , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Acetaminophen/toxicity , Acetaminophen/metabolism , Interleukin-6/metabolism , DNA Helicases/metabolism , Interleukin-4/metabolism , Stress Granules , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Macrophages/metabolism , Nucleotidyltransferases/metabolism , Chemical and Drug Induced Liver Injury/metabolism , RNA, Messenger/metabolism , Mesenchymal Stem Cells/metabolism
4.
Front Oncol ; 12: 922945, 2022.
Article in English | MEDLINE | ID: mdl-35769713

ABSTRACT

Autotaxin (ATX) is a secreted glycoprotein and catalyzes the hydrolysis of lysophosphatidylcholine to lysophosphatidic acid (LPA), a growth factor-like signaling phospholipid. ATX has been abundantly detected in the culture medium of various cancer cells, tumor tissues, and serum or plasma of cancer patients. Biological actions of ATX are mediated by LPA. The ATX-LPA axis mediates a plethora of activities, such as cell proliferation, survival, migration, angiogenesis, and inflammation, and participates in the regulation of various physiological and pathological processes. In this review, we have summarized the physiological function of ATX and the ATX-LPA axis in liver cancer, analyzed the role of the ATX-LPA axis in tumorigenesis and metastasis, and discussed the therapeutic strategies targeting the ATX-LPA axis, paving the way for new therapeutic developments.

5.
World J Gastrointest Oncol ; 13(12): 2029-2037, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-35070039

ABSTRACT

With the advance of genome engineering technology, chimeric antigen receptors (CARs)-based immunotherapy has become an emerging therapeutic strategy for tumors. Although initially designed for T cells in tumor immunotherapy, CARs have been exploited to modify the function of natural killer (NK) cells against a variety of tumors, including hepatocellular carcinoma (HCC). CAR-NK cells have the potential to sufficiently kill tumor antigen-expressing HCC cells, independent of major histocompatibility complex matching or prior priming. In this review, we summarize the recent advances in genetic engineering of CAR-NK cells against HCC and discuss the current challenges and prospects of CAR-NK cells as a revolutionary cellular immunotherapy against HCC.

6.
Cancer Cell Int ; 20: 332, 2020.
Article in English | MEDLINE | ID: mdl-32699531

ABSTRACT

BACKGROUND: Fatty acid synthase (FASN) is highly expressed in various types of cancer and has an important role in carcinogenesis and metastasis. To clarify the mechanisms of FASN in liver cancer invasion and metastasis, the FASN protein interaction network in liver cancer was identified by targeted proteomic analysis. METHODS: Wound healing and Transwell assays was performed to observe the effect of FASN during migration and invasion in liver cancer. Isobaric tags for relative and absolute quantitation (iTRAQ)-based mass spectrometry were used to identify proteins interacting with FASN in HepG2 cells. Differential expressed proteins were validated by co-immunoprecipitation, western blot analyses and confocal microscopy. Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to demonstrate the mechanism of FASN regulating metastasis. RESULTS: FASN knockdown inhibited migration and invasion of HepG2 and SMMC7721 cells. A total of, 79 proteins interacting with FASN were identified. Additionally, gene ontology term enrichment analysis indicated that the majority of biological regulation and cellular processes that the FASN-interacting proteins were associated with. Co-precipitation and co-localization of FASN with fascin actin-bundling protein 1 (FSCN1), signal-induced proliferation-associated 1 (SIPA1), spectrin ß, non-erythrocytic 1 (SPTBN1) and CD59 were evaluated. Knockdown of FASN in liver cancer reduced the expression of FSCN1, SIPA1, SPTBN1 and CD59. Furthermore, inhibition of FASN, FSCN1 or SPTBN1 expression in liver cancer resulted in alterations of epithelial-mesenchymal transition (EMT)-associated markers E-cadherin, N-cadherin, vimentin and transcription factors, Snail and Twist, at the mRNA level, and changes in matrix metallopeptidase (MMP)-2 and MMP-9 protein expression. CONCLUSION: The results suggested that the FASN-interacting protein network produced by iTRAQ-based proteomic analyses may be involved in regulating invasion and metastasis in liver cancer by influencing EMT and the function of MMPs.

7.
Immunobiology ; 225(3): 151934, 2020 05.
Article in English | MEDLINE | ID: mdl-32173150

ABSTRACT

NK-92 cell line has been used as anti-tumor cytotoxic effector cells in immunotherapy. Leucine-rich repeats and calponin homology domain containing 1 (LRCH1) is a novel gene of which the function is unclear. In the present study, we investigated the role of LRCH1 in NK-92 cell cytotoxicity. LRCH1 was ablated in NK-92 cells through CRISP-Cas9-mediated knockout. LRCH1 knockout did not influence the basal behavior of NK-92 cells such as cell survival, expression of natural cytotoxicity receptors, and proliferation. However, upon the contact with tumor cells, LRCH1 knockout promoted NK-92 cell cytotoxicity to tumor cells. Besides, LRCH1 knockout increased the production of cytotoxic mediators such as IFN-γ, TNF-α, IL-2, and granzyme B in NK-92 cells after tumor cell contact. Similarly, LRCH1 knockout increased the production of cytokines and granzyme B upon NKp30 engagement. Further experiments revealed that LRCH1 knockout enhanced the activation of Src and Lck kinase which are important for natural killer cell cytotoxicity. The in vivo assay confirmed the up-regulation of the tumoricidal activity of LRCH1-/- NK-92 cells, as demonstrated by more robust tumor cell killing. Importantly, human primary natural killer cells exhibited a similar increase in the production of IFN-γ and TNF-α when LRCH1 was knocked out. In conclusion, our study revealed the role of LRCH1 as a negative regulator of NK-92 cell cytotoxicity.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Microfilament Proteins/metabolism , Protein Interaction Domains and Motifs , Signal Transduction , src-Family Kinases/metabolism , Biomarkers , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Line , Cytokines/metabolism , Humans , Leucine/chemistry , Leucine/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Repetitive Sequences, Nucleic Acid , Calponins
8.
Int J Oncol ; 54(3): 1086-1098, 2019 03.
Article in English | MEDLINE | ID: mdl-30628664

ABSTRACT

Globally, gastric cancer is the fifth most common malignancy, with high rates of incidence and mortality. The high mortality rate and poor prognosis of gastric cancer are closely associated with its profound invasiveness, high incidence of metastasis, rapid proliferation, and high rate of recurrence. Previous studies have confirmed that stathmin (STMN) has an important role in the occurrence, development and prognosis of gastric cancer. However, the detailed mechanisms by which STMN affects these processes remain unclear. The aim of the present study was to determine how STMN promotes invasion, migration and proliferation in gastric cancer tumor cells. The results of immunohistochemistry indicated that STMN is overexpressed in stomach neoplasm tissues, and that it is associated with migration, invasion, proliferation and anti­apoptotic states of gastric cancer cells. The secretory proteins of gastric cancer cells with or without STMN knockdown were further analyzed using the isobaric tags for relative and absolute quantitation method to identify differentially expressed proteins verified by reverse transcription­quantitative polymerase chain reaction and western blot analysis. Inhibition of STMN decreases the levels of clusterin, cystatin C and matrix metalloproteinases, followed by inhibiting the protein kinase B and signal transducer and activation of transcription activation. These findings suggest that STMN could be a promising therapeutic target for gastric cancer.


Subject(s)
Clusterin/metabolism , Gene Silencing , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Stathmin/genetics , Stomach Neoplasms/pathology , Aged , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Clusterin/genetics , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-akt/genetics , STAT3 Transcription Factor/genetics , Stathmin/metabolism , Stomach Neoplasms/metabolism
9.
Cell Physiol Biochem ; 48(2): 741-752, 2018.
Article in English | MEDLINE | ID: mdl-30025407

ABSTRACT

BACKGROUND/AIMS: C reactive protein (CRP) levels are elevated in many diseases, including malignant tumors and cardiovascular disorders. In this study, the protein interaction network for CRP was evaluated to determine the importance of CRP and its interacting proteins in the molecular pathogenesis of hepatocellular carcinoma (HCC). METHODS: Isobaric tags for relative and absolute quantitation (iTRAQ) and mass spectrometry were used to identify CRP interacting proteins in SMMC7721 cells. Moreover, Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to evaluate enriched genes and pathways for differentially expressed genes using DAVID and WebGestalt. Co-immunoprecipitation and western blot analyses were employed to assess interactions between CRP and KRT8, ANXA2, ENO2, and HSP90B1. RESULTS: In total, 52 proteins that interact with CRP were identified. A GO analysis suggested that most of the interacting proteins were involved in CRP complexes and regulated metabolic processes. A KEGG pathway analysis suggested that most CRP-interacting proteins contribute to the TRAIL signaling pathway, Class I PI3K/Akt signaling pathway, plasma membrane estrogen receptor signaling, Nectin adhesion pathway, and S1P1 pathway. Immunoprecipitation and western blot analyses revealed interactions between CRP and KRT8, ANXA2, ENO2, and HSP90B1. CONCLUSIONS: iTRAQ based proteomic profiling revealed the network of CRP interacting proteins. This network may activate the PI3K/Akt signaling pathway, thereby contributing to the pathogenesis of HCC.


Subject(s)
C-Reactive Protein/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Proteomics , Annexin A2/metabolism , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/genetics , Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling , Humans , Keratin-8/metabolism , Liver Neoplasms/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Nectins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism
10.
Cell Physiol Biochem ; 45(2): 744-760, 2018.
Article in English | MEDLINE | ID: mdl-29414802

ABSTRACT

BACKGROUND/AIMS: Hepatitis B virus (HBV) infection is a major cause of cirrhosis and hepatocellular carcinoma. Therefore, we aimed to obtain further information on HBV pathogenesis, and to search for novel putative molecules for anti-HBV therapy. METHODS: We utilized Isobaric Tags for Relative and Absolute Quantitation (iTRAQ) to identify the secretory proteins that are differentially expressed in the HBV DNA-transfected HepG2.2.15 cell line and its parental HepG2 cell line. Immunohistochemistry (IHC) was employed to assess the clinical relevance of the observations. Small interfering (si)RNA-based silencing transfection methods were carried out to study the function of ENPP2. RESULTS: Totally, 133 unique proteins were identified as differentially expressed in HepG2.2.15 cell line compared with HepG2 cell line. Ectonucleotide pyrophosphatase/phosphodiesterase family member 2 precursor (ENPP2) is one of the most significantly up-regulated secretory proteins associated with HBV replication. This differential expression of ENPP2 was further validated by real-time quantitative RT-PCR, Western Blot and immunohistochemical analysis. To study the function of ENPP2, we knockdown ENPP2 expression in HepG2.2.15 cell line by RNA interference. ENPP2 silencing increased HBV replication approximately 2.3-fold by enhancing, via the type I IFN signaling pathway, HBV cccDNA (covalently closed circular DNA) translation into viral RNA. Moreover, attenuation of ENPP2 expression inhibited both the invasion and migration ability of hepatoma cells in vitro via interacting with the molecules in the tumor microenvironment. CONCLUSION: Our study demonstrates that ENPP2 may be a novel anti-HBV target and indicate that suppression of its expression may inhibit the invasion and migration ability of hepatoma cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepatitis B virus/physiology , Liver Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Movement , DNA, Viral/physiology , Gene Expression Regulation , Hep G2 Cells , Humans , Interferon Type I/metabolism , Liver Neoplasms/blood , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Middle Aged , Phosphoric Diester Hydrolases/blood , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Proteoglycans/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Virus Replication
11.
Mol Med Rep ; 17(3): 3481-3488, 2018 03.
Article in English | MEDLINE | ID: mdl-29286136

ABSTRACT

Despite the use of adjuvant therapies, the cumulative proportion of live births remains at ~40%. Accumulating data show that low pregnancy rates, even in the presence of high fertility rates, are due to implantation failure. The present study aimed to identify and construct a profile of proteins that react with preimplantation factor (PIF) and to provide an understanding into the molecular mechanisms by which PIF promotes trophoblast invasion. Cytoplasmic proteins were immunoprecipitated with biotin­labeled synthetic PIF or intralipid and scrambled PIF (PIFscr). The protein profiles were analyzed using isobaric tags for relative and absolute quantification coupled with mass spectrometry. Immunoprecipitation and western blot analyses were used to assess the interactions between PIF and myosin heavy chain 10 (MYH10) and heat shock protein family D1. Small interfering RNA­based silencing was performed to examine the function of MYH10. In the results of the present study, 21 proteins were identified with interactions with PIF. The immunoprecipitation and western blot analyses revealed an interaction between PIF and MYH10. Silencing of the expression of MYH10 in HEC­1­B cells significantly attenuated cell migration and invasion capacities. These data support the conclusion that MYH10­mediated cell migration and invasion act in conjunction with PIF to promote the trophoblast invasion procedure.


Subject(s)
Embryo Implantation/drug effects , Peptides/pharmacology , Proteome/metabolism , Cell Line , Cell Movement/drug effects , Chromatography, High Pressure Liquid , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , HEK293 Cells , Humans , Mass Spectrometry , Myosin Heavy Chains/antagonists & inhibitors , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIB/antagonists & inhibitors , Nonmuscle Myosin Type IIB/genetics , Nonmuscle Myosin Type IIB/metabolism , Proteome/drug effects , Proteomics , RNA Interference , RNA, Small Interfering/metabolism , Tubulin/genetics , Tubulin/metabolism
12.
Oncotarget ; 8(61): 104258-104271, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262638

ABSTRACT

Gastric cancer (GC) is a globally occurring malignancy that is characterized by a high mortality rate due to a high tendency to metastasize and poor prognoses. Sorcin, as known as SRI, a soluble resistance-related calcium-binding protein, plays a significant role in multidrug resistance. Sorcin is related to the migration and invasion of cancer cells. However, the mechanism remains unclear. Here, we used immunohistochemistry to confirm that the expression of sorcin in cancer tissues is higher than that in the adjacent normal tissues. The wound healing and transwell results indicate that sorcin can induce migration and invasion of GC cells. To explore the role of sorcin in GC metastasis, isobaric tags for relative and absolutely quantitation (iTRAQ) were used to examine cells with and without sorcin knockdown to identify the differentially expressed proteins (DEPs). The results were evaluated via RT-PCR and western blot to confirm the ITRAQ data. Inhibition of sorcin expression can down- regulate the expression of CTSZ, MMP2, MMP9 and p-STAT3 followed by suppression of tumor growth and metastasis. Together, we concluded that sorcin has a oncogenic activity via inducing tumor growth and metastasis, leading to development of therapeutic treatments for GC.

13.
Dig Liver Dis ; 49(7): 780-788, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28377286

ABSTRACT

BACKGROUND: Liver fibrosis can lead to cirrhosis and hepatocellular carcinoma if not treated in the early stages. The molecular mechanisms of the pathogenesis of hepatic fibrosis remain unclear. AIM: To identify the molecules involved in the pathogenesis of liver fibrosis and to investigate the potential effect and mechanism of Annexin A2 up-regulation during liver fibrosis progression. METHODS: Twenty Sprague-Dawley rats were divided into two groups: the carbon tetrachloride (CCl4)-induced liver fibrosis group and the normal control group. Hematoxylin and eosin staining or Masson Trichrome staining and enzyme-linked immunosorbent assay were applied to assess the degree of liver damage and fibrosis in rats with CCl4-induced liver fibrosis. Liver tissue protein profiles were analyzed using iTRAQ and mass spectrometry. RT-PCR and western blotting analyses were employed to validate differentially expressed proteins. Small interfering RNA-based silencing was performed to study the function of Annexin A2. RESULTS: Twelve weeks after CCl4 injection, significant body weight changes and liver injury and liver fibrosis were observed in rats. In addition, 130 proteins were differentially expressed in the liver fibrosis group. Overexpression of Annexin A2 was confirmed by RT-PCR and Western blotting analysis. Silencing of Annexin A2 expression in HepG2 and LX-2 cells significantly reduced the secretion of von Willebrand factor (vWF). CONCLUSION: Annexin A2 promotes liver fibrosis by mediating vWF secretion, which can be used to mitigate the progression of liver fibrosis.


Subject(s)
Annexin A2/metabolism , Liver Cirrhosis, Experimental/metabolism , von Willebrand Factor/metabolism , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Silencing , Liver Cirrhosis, Experimental/etiology , Mass Spectrometry , Protein Binding , RNA, Small Interfering , Random Allocation , Rats , Rats, Sprague-Dawley , Up-Regulation
14.
Int J Oncol ; 50(4): 1289-1298, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28350119

ABSTRACT

Hepatocellular carcinoma (HCC) is one of most common malignant cancers and is the second leading cause of cancer related deaths. The prognosis and survival of patients are closely related to the degree of tumor metastasis. The mechanism of HCC metastasis is still unclear. In the present study, we investigated the molecular mechanism of C-reaction protein in promoting migration and invasion of hepatocellular carcinoma cells in vitro. We estimated that CRP is overexpressed in liver cancer tissues and that it promotes invasion and metastasis of HCC in vitro. In the present study, we employed iTRAQ-based mass spectrometry to analyze the HepG2 secretory proteins of CRP siRNA-treated cells and negative control siRNA-treated cells. We identified 109 differentially expressed proteins after silencing CRP, of which 45 were upregulated and 64 were downregulated. Some of the differentially expressed proteins were confirmed by western blot analysis and real-time quantitative PCR. Furthermore, we found that knockdown of CRP substantially abrogates HIF-1α expression levels, the luciferase activity of HIF-1α and ERK and Akt phosphorylation in HepG2 cells. The present study provides a novel mechanism by which CRP promotes the proliferation, migration, invasion and metastasis of hepatocellular carcinoma cells. Inhibition of CRP suppressed migration, invasion and healing of hepatoma carcinoma cells by decreasing HIF-1α activity and CTSD.

15.
Int J Oncol ; 50(3): 883-892, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28197637

ABSTRACT

Hepatocellular carcinoma is the second most common cause of cancer-related deaths worldwide. Due to a high propensity to metastasize, active angiogenesis and rapid proliferation, recurrence and poor prognosis are major obstacles for treatment and cure of this disease. However, the detailed mechanisms of how fatty acid synthase (FASN) promotes migration, invasion and healing in tumor cells remain unclear. In the present study, the previous results that FASN was expressed higher in cancer samples than in non-cancerous samples, and influenced migration, invasion of hepatoma carcinoma cells, were verified by immunohistochemistry, tissue microarrays, Transwell assay and wound healing assay. The secretory proteins of hepatocellular carcinoma cells with or without FASN knockdown were analyzed using the isobaric tags for relative and absolute quantitation (iTRAQ) method to identify differentially expressed proteins (DEPs). The DEPs were verified by RT-PCR and western blot analysis, and were consistent with the iTRAQ results. Inhibition of FASN can decrease the levels of IGFBP1, and the expression, activity, and ubiquitination of HIF-1α. Inhibition of FASN can suppress migration, invasion and healing of hepatoma carcinoma cells by decreasing HIF-1α, and IGFBP1.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement/genetics , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Neoplasm Invasiveness/genetics , RNA Interference , RNA, Small Interfering/genetics
16.
Oncotarget ; 8(3): 4549-4562, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27999186

ABSTRACT

BACKGROUND AND AIMS: Hepatitis B virus (HBV) infection is a major risk factor for liver cirrhosis and hepatocellular carcinoma (HCC). To gain a better understanding of the pathogenesis of HBV infection, this study aimed to investigate the differentially expressed proteins (DEPs) in liver tissues from patients with chronic hepatitis B (CHB) infection. RESULTS: Seventy-one DEPs were identified. Overexpression of multi-drug resistance protein 1 (MDR1) was validated by RT-qPCR and western blot analyses. Moreover, its expression was increased at both the mRNA and protein levels in response to overexpression of HBV large surface protein (LHBs). Furthermore, screening of transcription factors suggested the possible involvement of hypoxia-inducible factor 1α (HIF-1α) in the interaction between LHBs and MDR1. The function of HIF-1α in the MDR1 activation was confirmed by EMSA and reporter gene analyses. MATERIALS AND METHODS: Liver samples from CHB patients and controls without HBV infection were collected and subjected to isobaric tags for relative and absolute quantitation (iTRAQ) and mass spectrometric analysis. CONCLUSIONS: These results imply that LHBs, in association with HIF-1α, induces MDR1 overexpression, which may contribute to the pathogenic changes in CHB infection.


Subject(s)
Carcinoma, Hepatocellular/virology , Hepatitis B, Chronic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/virology , Viral Envelope Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Adult , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatitis B virus/metabolism , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/genetics , Humans , Liver/metabolism , Liver/virology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mass Spectrometry , Middle Aged , Proteomics/methods , Young Adult
17.
Int J Oncol ; 47(5): 1932-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398045

ABSTRACT

Gastric cancer (GC) is now one of the most common malignancies with a relatively high incidence and high mortality rate. The prognosis is closely related to the degree of tumor metastasis. The mechanism of metastasis is still unclear. Proteomics analysis is a powerful tool to study and evaluate protein expression in tumor tissues. In the present study, we collected 15 gastric cancer and adjacent normal gastric tissues and used the isobaric tags for relative and absolute quantitation (iTRAQ) method to identify differentially expressed proteins. A total of 134 proteins were differentially expressed between the cancerous and non-cancerous samples. Azurocidin 1 (AZU1), CPVL, olfactomedin 4 (OLFM4) and Villin 1 (VIL1) were upregulated and confirmed by western blot analysis, real-time quantitative PCR and immunohistochemical analyses. These results were in accordance with iTRAQ. Furthermore, silencing the OLFM4 expression suppressed the migration, invasion and proliferation of the GC cells in vitro. The present study represents a successful application of the iTRAQ method in analyzing the expression levels of thousands of proteins. Overexpression of OLFM4 in gastric cancer may induce the development of gastric cancer. Overall, suppression of OLFM4 expression may be a promising strategy in the development of novel cancer therapeutic drugs.


Subject(s)
Biomarkers, Tumor/biosynthesis , Granulocyte Colony-Stimulating Factor/biosynthesis , Proteomics , Stomach Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Granulocyte Colony-Stimulating Factor/genetics , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology
18.
Int J Oncol ; 47(2): 543-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26058824

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most aggressive cancers worldwide and is associated with the high rates of morbidity and mortality. α-fetoprotein (AFP) is common used in diagnosis of HCC; however, a growing body of research is questioning the diagnostic power of AFP. There is, therefore, an urgent need to develop additional novel non-invasive techniques for the early diagnosis of HCC, particularly for patients with AFP-negative [AFP(-)] HCC. Accordingly, in the present study, we employed iTRAQ-based mass spectro-metry to analyze the plasma proteins of subjects with AFP(-) HBV-related HCC, AFP(+) HBV-related HCC and non-malignant cirrhosis. We identified 14 aberrantly expressed proteins specific to the HCC patients, including 10 upregulated and 4 downregulated proteins. We verified C-reactive protein (CRP) overexpression by ELISA and immunohistochemical staining of clinical samples. Per ROC curve analyses, CRP was positive in 73.3% of patients with HBV-related HCC, and CRP overexpression had significant diagnostic power for AFP(-) HBV-related HCC. Furthermore, we found that silencing CRP caused a >2-fold decease in HBV replication. Additionally, we determined that this reduction in HBV replication involved the interferon-signaling pathway. However, silencing CRP also promoted HCC invasion and migration in vitro. In conclusion, we demonstrated that CRP can serve as a diagnostic biomarker for AFP(-) HBV-related HCC.


Subject(s)
Biomarkers, Tumor/blood , C-Reactive Protein/metabolism , Carcinoma, Hepatocellular/virology , Hepatitis B/complications , Liver Neoplasms/virology , alpha-Fetoproteins/deficiency , Adolescent , Adult , Aged , Carcinoma, Hepatocellular/blood , Cell Line, Tumor , Early Detection of Cancer , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatitis B/blood , Humans , Liver Neoplasms/blood , Male , Mass Spectrometry/methods , Middle Aged , ROC Curve , Virus Replication , Young Adult
19.
J Cell Biochem ; 116(7): 1431-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25648846

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a common disease in the southern provinces of China with a poor prognosis. To better understand the pathogenesis of NPC and identify proteins involved in NPC carcinogenesis, we applied iTRAQ coupled with two-dimensional LC-MS/MS to compare the proteome profiles of NPC tissues and the adjacent non-tumor tissues. We identified 54 proteins with differential expression in NPC and the adjacent non-tumor tissues. The differentially expressed proteins were further determined by RT-PCR and Western blot analysis. In addition, the up-regulation of HSPB1, NPM1 and NCL were determined by immunohistochemistry using tissue microarray. Functionally, we found that siRNA mediated knockdown of NPM1 inhibited the migration and invasion of human NPC CNE1 cell line. In summary, this is the first study on proteome analysis of NPC tissues using an iTRAQ method, and we identified many new differentially expressed proteins which are potential targets for the diagnosis and therapy of NPC.


Subject(s)
Gene Expression Regulation, Neoplastic , Nasopharyngeal Neoplasms/metabolism , Proteomics/methods , Carcinoma , Cell Line, Tumor , Cell Movement , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Nucleolin
20.
Int J Oncol ; 46(4): 1748-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25633909

ABSTRACT

Cervical cancer is the seventh most common cancer overall and the third among females. To obtain systematic insight into the protein profile that participates in cervical tumor oncogenesis and improve the current target therapies, iTRAQ labeling and NanoLC-MS/MS analysis were utilized to detect differentially expressed proteins in cervical cancer. As a result, 3,647 proteins were identified, among which the expression levels of 294 proteins in cervical cancer samples were distinct from the paired non-tumor samples. Further validation of the differentially expressed proteins, including G6PD, ALDH3A1, STAT1 and HSPB1, was carried out via qRT-PCR, western blot analysis and tissue microarray. Functional analysis of one of the highly expressed proteins, G6PD, was performed using RNA interference. Attenuated G6PD expression reduced the capacity of HeLa cells to migrate and invade in vitro. Our investigation complemented the understanding of cervical cancer progression. Furthermore, the present study supports the notion that suppressing the expression of G6PD may be a promising strategy in developing novel cancer therapeutic drugs.


Subject(s)
Gene Expression Profiling/methods , Glucosephosphate Dehydrogenase/metabolism , Isotope Labeling/methods , Proteomics/methods , Uterine Cervical Neoplasms/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glucosephosphate Dehydrogenase/genetics , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , HeLa Cells , Heat-Shock Proteins , Humans , Molecular Chaperones , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Tandem Mass Spectrometry/methods , Tissue Array Analysis/methods , Uterine Cervical Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...