Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
PLoS One ; 11(2): e0148381, 2016.
Article in English | MEDLINE | ID: mdl-26839961

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the predominant pathotype of esophageal carcinoma (EC) in China, especially in Henan province, with poor prognosis and limited 5-year survival rate. Cellular retinoic acid binding protein 2 (CRABP2) is a member of the retinoic acid (RA) and lipocalin/cytosolic fatty-acid binding protein family and plays a completely contrary role in tumorigenesis through the retinoid signaling pathway, depending on the nuclear RA receptors (RAR) and PPARbeta/delta receptors. Presently, the biological role of CRABP2 in the development of ESCC has never been reported. Here, we firstly evaluated the expression of CRABP2 at both mRNA and protein levels and showed that it was remarkably downregulated in clinical ESCC tissues and closely correlated with the occurrence position, pathology, TNM stage, size, infiltration depth and cell differentiation of the tumor. Additionally, the biological function assays demonstrated that CRABP2 acted as a tumor suppressor in esophageal squamous carcinogenesis by significantly inhibiting cell growth, inducing cell apoptosis and blocking cell metastasis both in vitro and in vivo. All in all, our finding simplicate that CRABP2 is possibly an efficient molecular marker for diagnosing and predicting the development of ESCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Retinoic Acid/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Apoptosis/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Retinoic Acid/genetics , Tumor Suppressor Proteins/genetics
2.
Dev Growth Differ ; 57(8): 581-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26449203

ABSTRACT

Cellular retinoic acid binding protein 2 (CRABP2) is essential for myoblast differentiation, however, little is known about its role in osteogenic differentiation. This study mainly aims to explore the biological functions and the underlying molecular mechanisms of CRABP2 in osteogenesis. Using quantitative polymerase chain reaction and western blot assays, we found that the expression of CRABP2 at both mRNA and protein levels were downregulated during osteogenesis. Furthermore, CRABP2 knockdown displayed significant changes in the cell phenotype and the actin filaments (F-actin) polymerization in C2C12 cells treated with BMP2. Moreover, the western blotting of osteogenic differentiation biomarkers, alkaline phosphatase (ALP) staining and Alizarin red staining showed that CRABP2 dramatically inhibited osteogenic differentiation. The following investigation of molecular mechanisms implicated that CARBP2 specifically interacted with LIMK1, a key factor in acin cytoskeletal rearrangements in osteogenesis, to interrupt its activity and stability in an ubiquitin-proteasome pathway to prevent C2C12 cells from osteogenic differentiation in response to BMP2. Above all, our data suggest a novel function of CRABP2 in regulating actin remodeling and osteogenic differentiation via LIMK1, thus presenting a possible molecular target for promoting the osteogenic differentiation in bone degenerative diseases.


Subject(s)
Lim Kinases/metabolism , Receptors, Retinoic Acid/metabolism , Actins/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Lim Kinases/genetics , Mice , Osteogenesis/genetics , Osteogenesis/physiology , Protein Binding , Receptors, Retinoic Acid/genetics
3.
Int J Clin Exp Pathol ; 7(8): 4817-26, 2014.
Article in English | MEDLINE | ID: mdl-25197352

ABSTRACT

DNA damage response and repair are carried out by certain proteins following damage by environmental clastogens, such as ionizing radiation and reactive oxygen species. It has been reported that many carcinomas that are characterized by resistance to chemotherapy and poor outcomes show dysfunction of these proteins. Chromobox homologue 8 (CBX8), a member of the polycomb group of proteins, has been identified as a factor that protects tumor cells from the detrimental effects of ionizing radiation (IR) or hydrogen peroxide (H2O2). In this study, we found that CBX8 was up-regulated in esophageal carcinoma tissues compared with adjacent non-cancerous tissues (P<0.01) and correlated with TNM stage in esophageal squamous cell carcinoma patients. Depletion of CBX8 decreased cell proliferation both in vitro and in vivo and increased the phosphorylation levels of p21, Wee1, and CHK1, which result in cyclin-dependent kinase inhibition and cell-cycle delay. CBX8 depletion also led to accumulation of spontaneous DNA damage and raised the sensitivity of tumor cells to IR or H2O2. We also found that the total level of CBX8 in the cells was increased after treating tumor cells with clastogens. In addition, our data showed that decreased CBX8 expression was accompanied by the reduction of EZH2 and EED, which have been reported to participate in DNA damage repair. Collectively, CBX8 might emerge as an oncogene for promoting the proliferation of tumor cells and raising the resistance of neoplasms to chemotherapy.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/pathology , Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/pathology , Polycomb Repressive Complex 1/metabolism , Animals , Blotting, Western , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Polycomb Repressive Complex 1/genetics , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection
4.
Carcinogenesis ; 35(7): 1643-51, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24710624

ABSTRACT

Neurotrophin receptor-interacting melanoma antigen-encoding gene homolog (NRAGE) is generally recognized as a tumor suppressor as it induces cell apoptosis and suppresses cell metastasis. However, it has recently been reported that NRAGE is overexpressed in lung cancer, melanoma and colon cancer, implicating a complicated role of NRAGE as we have expected. In the study, we aim to elucidate the functional roles and molecular mechanisms of NRAGE in esophageal carcinoma. We found that both NRAGE mRNA and protein were significantly overexpressed in esophageal tumor tissues. Consistently, both in vivo and in vitro analyses demonstrated that knockdown of NRAGE apparently inhibited cell growth, and cell cycle analysis further demonstrated that NRAGE knockdown cells were mainly arrested in G2M cell phase, accompanied with an apparent reduction of S phase. In the process of exploring molecular mechanisms, we found that either knockdown in vitro or knockout in vivo of NRAGE reduced proliferating cell nuclear antigen (PCNA) protein, expression of which could completely rescue the inhibited proliferation in NRAGE defective cells. Furthermore, NRAGE physically interacted with PCNA in esophageal cancer cells through DNA polymerase III subunit, and knockdown of NRAGE facilitated PCNA K48-linked polyubiquitination, leading PCNA to the proteasome-dependent degradation and a ubiquitin-specific protease USP10 was identified to be a key regulator in the process of K48 polyubiquitination in NRAGE-deleted cells. In conclusion, our study highlights a unique role of NRAGE and implies that NRAGE is likely to be an attractive oncotarget in developing novel genetic anticancer therapeutic strategies for esophageal squamous cell carcinomas.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Proliferation , Esophageal Neoplasms/pathology , Esophagus/metabolism , Neoplasm Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism , Animals , Antigens, Neoplasm/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cohort Studies , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lymphatic Metastasis , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Staging , Proliferating Cell Nuclear Antigen/genetics , Proteasome Endopeptidase Complex/genetics , Proteolysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Xenograft Model Antitumor Assays
5.
Cell Signal ; 26(5): 1048-59, 2014 May.
Article in English | MEDLINE | ID: mdl-24468535

ABSTRACT

Emerging studies have revealed that Malat1 is overexpressed in many malignant diseases, including liver cancer, and contributes to enhancing cell migration or facilitating proliferation. However, the mechanism underlying its regulation has largely remained elusive. Here, we characterised the oncoprotein Yes-associated protein (YAP), which up-regulated metastasis-associated lung adenocarcinoma transcript 1 (Malat1) expression at both transcriptional and post-transcriptional levels, whereas serine/arginine-rich splicing factor 1 (SRSF1) played an opposing role. SRSF1 inhibited YAP activity by preventing its co-occupation with TCF/ß-catenin on the Malat1 promoter. In contrast, overexpression of YAP impaired the nuclear retention of both SRSF1 and itself via an interaction with Angiomotin (AMOT). This effect removed the inhibitory role of SRSF1 on Malat1 in the nucleus. Furthermore, higher expression of YAP was consistent with a lower SRSF1 nuclear accumulation in human liver cancer tissues. We also revealed that overexpression of YAP combined with a knockdown of SRSF1 resulted in conspicuously enhanced transwell cell mobility, accelerated tumour growth rate, and loss of body weight in a tail vein-injected mouse models. Taken together, these data provided a novel mechanism underlying the balance between SRSF1, YAP and Malat1 and uncovered a new role of YAP in regulating long non-coding RNA (lncRNA). Thus, disrupting the interaction between YAP and SRSF1 may serve as a crucial therapeutic method in liver cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Angiomotins , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , HEK293 Cells , Hep G2 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Mice , Mice, Nude , Microfilament Proteins , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Transcription Factor 4 , Transcription Factors/metabolism , Transcription, Genetic , Transplantation, Heterologous , Up-Regulation , YAP-Signaling Proteins , beta Catenin/metabolism
6.
FEBS Lett ; 587(24): 3921-7, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24211253

ABSTRACT

Mitogen-activated protein kinase kinase 1 (MAP2K1/MEK1) as well as Yes-associated protein (YAP), the downstream effector of Hippo signaling pathway, is linked to hepatocarcinogenesis. However, little is known about whether and how MEK1 interacts with YAP. In this study, we find that MEK1-YAP interaction is critical for liver cancer cell proliferation and maintenance of transformed phenotypes both in vitro and in vivo. Moreover, MEK1 and YAP proteins are closely correlated in human liver cancer samples. Mechanistically, inhibition of MEK1 by both PD98059 and U0126 as well as RNAi reduces beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC), which acts as a potential endogenous YAP protector.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MAP Kinase Kinase 1/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins , Cell Proliferation , HEK293 Cells , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Nude , Protein Binding/physiology , Tumor Cells, Cultured , Up-Regulation
7.
J Biol Chem ; 288(47): 33667-33681, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24089522

ABSTRACT

Tribbles homolog 2 (TRIB2) is critical for both solid and non-solid malignancies. Recently, TRIB2 was identified as a liver cancer-specific Wnt/ß-catenin signaling downstream target and is functionally important for liver cancer cell survival and transformation. TRIB2 functions as a protein that interacts with E3 ubiquitin ligases and thereby modulates protein stability of downstream effectors. However, the regulation underlying TRIB2 protein stability per se has not yet been reported. In this study, we found that TRIB2 was up-regulated and exhibited high stability in liver cancer cells compared with other cells. We performed a structure-function analysis of TRIB2 and identified a domain (amino acids 1-5) at the N terminus that interacted with the E3 ubiquitin ligase Smurf1 and was critical for protein stability. Deletion of this domain extended TRIB2 half-life time accompanied with a more significant malignant property compared with wild type TRIB2. Furthermore, Smurf1-mediated ubiquitination required phosphorylation of TRIB2 by p70 S6 kinase (p70S6K) via another domain (amino acids 69-85) that is also essential for correct TRIB2 subcellular localization. Mutation of Ser-83 diminished p70S6K-induced phosphorylation of TRIB2. Moreover, the high stability of TRIB2 may be due to the fact that both p70S6K and Smurf1 were down-regulated and negatively correlated with TRIB2 expression in both liver cancer tissues and established liver cancer cell lines. Taken together, impaired phosphorylation and ubiquitination by p70S6K and Smurf1 increase the protein stability of TRIB2 in liver cancer and thus may be helpful in the development of diagnosis and treatment strategies against this malignant disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitination , Calcium-Calmodulin-Dependent Protein Kinases , Down-Regulation/genetics , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Phosphorylation/genetics , Protein Stability , Protein Structure, Tertiary , Protein Transport/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ubiquitin-Protein Ligases/genetics
8.
Biochem Biophys Res Commun ; 439(2): 167-72, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23994632

ABSTRACT

Yes-associated protein (YAP), the downstream effector of Hippo signaling pathway as well as c-Myc has been linked to hepatocarcinogenesis. However, little is known about whether and how YAP and c-Myc interacts with each other. In this study, we find YAP-c-Myc interaction is critical for liver cancer cell both in vitro and in vivo. Moreover, both c-Myc and YAP proteins are closely correlated in human liver cancer samples. Mechanistically, YAP promotes c-Myc transcriptional output through c-Abl. By contrast, c-Myc enhances protein expression independent of transcription. Taken together, our study uncovers a novel positive auto-regulatory feedback loop underlying the interaction between YAP and c-Myc in liver cancer, suggesting YAP and c-Myc links Hippo/YAP and c-Myc pathways, and thus may be helpful in the development of effective diagnosis and treatment strategies against liver cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver/pathology , Phosphoproteins/metabolism , Protein Interaction Maps , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Cycle Proteins , Cell Line, Tumor , Humans , Liver/metabolism , Mice , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
9.
Hepatology ; 58(3): 1011-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23532963

ABSTRACT

UNLABELLED: Yes-associated protein (YAP), the downstream effecter of the Hippo-signaling pathway as well as cyclic adenosine monophosphate response element-binding protein (CREB), has been linked to hepatocarcinogenesis. However, little is known about whether and how YAP and CREB interact with each other. In this study, we found that YAP-CREB interaction is critical for liver cancer cell survival and maintenance of transformative phenotypes, both in vitro and in vivo. Moreover, both CREB and YAP proteins are highly expressed in a subset of human liver cancer samples and are closely correlated. Mechanistically, CREB promotes YAP transcriptional output through binding to -608/-439, a novel region from the YAP promoter. By contrast, YAP promotes protein stabilization of CREB through interaction with mitogen-activated protein kinase 14 (MAPK14/p38) and beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC). Gain-of-function and loss-of-function studies demonstrated that phosphorylation of CREB by MAPK14/p38 at ser133 ultimately leads to its degradation. Such effects can be enhanced by BTRC through phosphorylation of MAPK14/p38 at Thr180/Tyr182. However, YAP negatively controls phosphorylation of MAPK14/p38 through inhibition of BTRC expression. CONCLUSION: There is a novel positive autoregulatory feedback loop underlying the interaction between YAP and CREB in liver cancer, suggesting that YAP and CREB form a nexus to integrate the protein kinase A, Hippo/YAP, and MAPK14/p38 pathways in cancer cells and thus may be helpful in the development of effective diagnosis and treatment strategies against liver cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phosphoproteins/metabolism , Animals , Cell Line, Tumor , Cell Survival/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Feedback, Physiological/physiology , Heterografts , Homeostasis/physiology , Humans , In Vitro Techniques , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 14/metabolism , Phosphorylation/physiology , Transcription Factors , YAP-Signaling Proteins , beta-Transducin Repeat-Containing Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...