Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Hum Vaccin Immunother ; 19(1): 2202127, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37128699

ABSTRACT

DNA vaccines containing only antigenic components have limited efficacy and may fail to induce effective immune responses. Consequently, adjuvant molecules are often added to enhance immunogenicity. In this study, we generated a tumor vaccine using a plasmid encoding NMM (NY-ESO-1/MAGE-A3/MUC1) target antigens and immune-associated molecules. The products of the vaccine were analyzed in 293 T cells by western blotting, flow cytometry, and meso-scale discovery electrochemiluminescence. To assess the immunogenicity obtained, C57BL/6 mice were immunized using the DNA vaccine. The results revealed that following immunization, this DNA vaccine induced cellular immune responses in C57BL/6 mice, as evaluated by the release of IFN-γ, and we also detected increases in the percentages of nonspecific lymphocytes, as well as those of antigen-specific T cells. Furthermore, immunization with the pNMM vaccine was found to significantly inhibit tumor growth and prolonged the survival of mice with B16-NMM+-tumors. Our data revealed that pNMM DNA vaccines not only confer enhanced immunity against tumors but also provide a potentially novel approach for vaccine design. Moreover, our findings provide a basis for further studies on vaccine pharmacodynamics and pharmacology, and lay a solid foundation for clinical application.


Subject(s)
Cancer Vaccines , Neoplasms , Vaccines, DNA , Mice , Animals , Mice, Inbred C57BL , Antigens, Neoplasm , Adjuvants, Immunologic , Immunity, Cellular
2.
BMC Cancer ; 18(1): 259, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29510676

ABSTRACT

BACKGROUND: Non-small-cell lung cancer (NSCLC) is characterized by abnormalities of numerous signaling proteins that play pivotal roles in cancer development and progression. Many of these proteins have been reported to be correlated with clinical outcomes of NSCLC. However, none of them could provide adequate accuracy of prognosis prediction in clinical application. METHODS: A total of 384 resected NSCLC specimens from two hospitals in Beijing (BJ) and Chongqing (CQ) were collected. Using immunohistochemistry (IHC) staining on stored formalin-fixed paraffin-embedded (FFPE) surgical samples, we examined the expression levels of 75 critical proteins on BJ samples. Random forest algorithm (RFA) and support vector machines (SVM) computation were applied to identify protein signatures on 2/3 randomly assigned BJ samples. The identified signatures were tested on the remaining BJ samples, and were further validated with CQ independent cohort. RESULTS: A 6-protein signature for adenocarcinoma (ADC) and a 5-protein signature for squamous cell carcinoma (SCC) were identified from training sets and tested in testing sets. In independent validation with CQ cohort, patients can also be divided into high- and low-risk groups with significantly different median overall survivals by Kaplan-Meier analysis, both in ADC (31 months vs. 87 months, HR 2.81; P <  0.001) and SCC patients (27 months vs. not reached, HR 9.97; P <  0.001). Cox regression analysis showed that both signatures are independent prognostic indicators and outperformed TNM staging (ADC: adjusted HR 3.07 vs. 2.43, SCC: adjusted HR 7.84 vs. 2.24). Particularly, we found that only the ADC patients in high-risk group significantly benefited from adjuvant chemotherapy (P = 0.018). CONCLUSIONS: Both ADC and SCC protein signatures could effectively stratify the prognosis of NSCLC patients, and may support patient selection for adjuvant chemotherapy.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Prospective Studies , Signal Transduction , Survival Rate , Tissue Array Analysis
3.
Cancer Invest ; 33(9): 469-75, 2015.
Article in English | MEDLINE | ID: mdl-26305602

ABSTRACT

This study aimed to investigate the expression of Friend leukemia virus integration 1 (Fli-1) and its correlation with the prognosis of endometrial cancer (EC). Thirty-two EC tissue samples were evaluated for Fli-1 expression using immunohistochemistry. Fli-1 showed significantly high expression in EC cells, followed by hyperplasia cells, and was negative in adjacent normal tissues. The high expression of Fli-1 was significantly associated with a high differentiation grade, mutated P53 expression, and histological subtype (p < .05). Downregulation of Fli-1 in AN3CN cells using RNA interference inhibited cell clone formation and proliferation but did not affect apoptosis and migration of the cells. This study provides the first evidence that Fli-1 expression gradually increases in parallel with disease progression, and its overexpression might predict poor prognosis in EC.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression/genetics , Microfilament Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Adult , Aged , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Middle Aged , Prognosis , RNA Interference/physiology , Trans-Activators
4.
Cell Rep ; 7(6): 1982-93, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24882011

ABSTRACT

CUEDC2, a CUE-domain-containing protein, modulates inflammation, but its involvement in tumorigenesis is still poorly understood. Here, we report that CUEDC2 is a key regulator of macrophage function and critical for protection against colitis-associated tumorigenesis. CUEDC2 expression is dramatically upregulated during macrophage differentiation, and CUEDC2 deficiency results in excessive production of proinflammatory cytokines. The level of CUEDC2 in macrophages is modulated by miR- 324-5p. We find that Cuedc2 KO mice are more susceptible to dextran-sodium-sulfate-induced colitis, and macrophage transplantation results suggest that the increased susceptibility results from the dysfunction of macrophages lacking CUEDC2. Furthermore, we find that Cuedc2 KO mice are more prone to colitis-associated cancer. Importantly, CUEDC2 expression is almost undetectable in macrophages in human colon cancer, and this decreased CUEDC2 expression is associated with high levels of interleukin-4 and miR-324-5p. Thus, CUEDC2 plays a crucial role in modulating macrophage function and is associated with both colitis and colon tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/immunology , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Female , Gene Expression Regulation , HeLa Cells , Humans , Macrophage Activation , Macrophages/immunology , Macrophages/pathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Transgenic , MicroRNAs/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction
5.
Zhonghua Yi Xue Za Zhi ; 93(14): 1099-103, 2013 Apr 09.
Article in Chinese | MEDLINE | ID: mdl-23902846

ABSTRACT

OBJECTIVE: To establish Gankyrin knocking down 4T1-luc cell model and detect the effects of Gankyrin expression on breast cancer metastasis. METHODS: 4T1-luc cells carrying shGankyrin construct were established by lentivirus infection and antibiotic screening. Western blotting and real-time PCR were used to check the expression levels of Gankyrin. In vivo imaging system was used to monitor the effects of Gankyrin knocked down on cell growth and tumor metastasis after the in situ implantation of Gankyrin knocking down 4T1-luc cells in BALB/c mice. RESULTS: The cell expression decreased at the protein and mRNA levels. Gankyrin mRNA expression in different shGankyrin 4T1-luc cells was respectively 4.9%, 25.1% and 69.8% versus the control cells. ShGankyrin#2 4T1-luc cells were chosen for in situ implantation into BAL/c mice because luminescent intensity was consistent with cell numbers. The photon flux of lung metastatic tumor induced by Gankyrin knocking down 4T1-luc cell was 3.02 × 10(6), while that of lung metastasis induced by control cells was 10.9 × 10(6). The differences between two groups were significant. In pathology, Gankyrin was detected positive in lung metastasis tumors induced by control group. However, Gankyrin was negative in the Gankyrin knockdown group. CONCLUSIONS: Lentivirus infection may be effectively used to establish Gankyrin knocking down 4T1-luc cell model. Because of its involvement in the in vivo pulmonary metastasis of breast cancers, Gankyrin should be a novel target for tumor therapy.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line , Cell Line, Tumor , Female , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis
6.
Proc Natl Acad Sci U S A ; 110(27): 11017-22, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776205

ABSTRACT

DNA damage triggers cell cycle arrest to provide a time window for DNA repair. Failure of arrest could lead to genomic instability and tumorigenesis. DNA damage-induced G1 arrest is generally achieved by the accumulation of Cyclin-dependent kinase inhibitor 1 (p21). However, p21 is degraded and does not play a role in UV-induced G1 arrest. The mechanism of UV-induced G1 arrest thus remains elusive. Here, we have identified a critical role for CUE domain-containing protein 2 (CUEDC2) in this process. CUEDC2 binds to and inhibits anaphase-promoting complex/cyclosome-Cdh1 (APC/C(Cdh1)), a critical ubiquitin ligase in G1 phase, thereby stabilizing Cyclin A and promoting G1-S transition. In response to UV irradiation, CUEDC2 undergoes ERK1/2-dependent phosphorylation and ubiquitin-dependent degradation, leading to APC/C(Cdh1)-mediated Cyclin A destruction, Cyclin-dependent kinase 2 inactivation, and G1 arrest. A nonphosphorylatable CUEDC2 mutant is resistant to UV-induced degradation. Expression of this stable mutant effectively overrides UV-induced G1-S block. These results establish CUEDC2 as an APC/C(Cdh1) inhibitor and indicate that regulated CUEDC2 degradation is critical for UV-induced G1 arrest.


Subject(s)
Carrier Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Membrane Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Adaptor Proteins, Signal Transducing , Anaphase-Promoting Complex-Cyclosome , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , G1 Phase Cell Cycle Checkpoints/radiation effects , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Phosphorylation/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ultraviolet Rays
7.
Stem Cells ; 31(4): 823-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23307608

ABSTRACT

Muscular dystrophies are a group of devastating diseases characterized by progressive muscle weakness and degeneration, with etiologies including muscle gene mutations and regenerative defects of muscle stem cells. Notch signaling is critical for skeletal myogenesis and has important roles in maintaining the muscle stem cell pool and preventing premature muscle differentiation. To investigate the functional impact of Notch signaling blockade in muscle stem cells, we developed a conditional knock-in mouse model in which endogenous Notch signaling is specifically blocked in muscle stem cell compartment. Mice with Notch signaling inhibition in muscle stem cells showed several muscular dystrophic features and impaired muscle regeneration. Analyses of satellite cells and isolated primary myoblasts revealed that Notch signaling blockade in muscle stem cells caused reduced activation and proliferation of satellite cells but enhanced differentiation of myoblasts. Our data thus indicate that Notch signaling controls processes that are critical to regeneration in muscular dystrophy, suggesting that Notch inhibitor therapies could have potential side effects on muscle functions.


Subject(s)
Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/physiology , Muscular Dystrophies/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Mice , Mice, Knockout , Muscle Development/genetics , Muscular Dystrophies/genetics , Myoblasts/cytology , Myoblasts/metabolism , Receptors, Notch/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
8.
BMC Med Genomics ; 5: 36, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22913272

ABSTRACT

BACKGROUND: Gankyrin was originally purified and characterized as the p28 component of the 26S proteasome, and later identified as an oncogenic protein in hepatocellular carcinomas (HCC). It has recently been found to be highly expressed in several other malignancies, and compelling evidence show gankyrin plays important roles in tumorigenesis. However, its mechanism of action remains unclear. METHODS: In order to further clarify the functions of gankyrin and better understand its molecular mechanisms, we generated a gankyrin null cell line, HCT116 gankyrin-/- , by targeted homologous recombination in human colon cancer cells, and then employed two-dimensional electrophoresis (2-DE) based proteomic approaches followed by MS identification to investigate alterations in the proteome due to the gankyrin knockout. Western blot and qRT-PCR assays were also used to examine the protein and mRNA levels of some identified proteins. RESULTS: Compared with wild-type control cells, gankyrin null cells were impaired in terms of their proliferation, migration and anchorage-independent growth. A total of 21 altered proteins were identified, which included 18 proteins that had not previously been reported to be related to gankyrin. Notably, eight metastasis-related proteins were identified. Western blot analyses confirmed that the changes in three examined proteins were consistent with 2-DE gel analysis. CONCLUSIONS: In summary, we have generated a useful cell tool to clarify the functions of gankyrin. Our proteomic data provide novel information to better understand the roles and underlying mechanisms by which gankyrin is involved in tumorigenesis and cancer metastasis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Deletion , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteomics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Cell Movement/genetics , Cell Proliferation , Colonic Neoplasms/metabolism , Gene Knockout Techniques , HCT116 Cells , Humans , Neoplasm Metastasis/genetics , Proteasome Endopeptidase Complex/deficiency , Proto-Oncogene Proteins/deficiency
9.
Oncologist ; 17(2): 220-32, 2012.
Article in English | MEDLINE | ID: mdl-22291092

ABSTRACT

The role of Notch signaling in cervical cancer is seemingly controversial. To confirm the function of Notch signaling in this type of cancer, we established a stable Notch1-activated cervical cancer HeLa cell line. We found that Notch1 activation resulted in apoptosis, cell cycle arrest, and tumor suppression. At the molecular level, we found that a variety of genes associated with cyclic AMP, G protein-coupled receptor, and cancer signaling pathways contributed to Notch1-mediated tumor suppression. We observed that the expression of somatostatin (SST) was dramatically induced by Notch1 signaling activation, which was accompanied by enhanced expression of the cognate SST receptor subtype 1 (SSTR1) and SSTR2. Certain genes, such as tumor protein 63 (TP63, p63), were upregulated, whereas others, such as B-cell lymphoma 2 (BCL-2), Myc, Akt, and STAT3, were downregulated. Subsequently, knockdown of Notch1-induced SST reversed Notch1-induced decrease of BCL-2 and increase of p63, indicating that Notch1-induced tumor suppression may be partly through upregulating SST signaling. Our findings support a possible crosstalk between Notch signaling and SST signaling. Moreover, Notch-induced SSTR activation could enhance SSTR-targeted cancer chemotherapy. Valproic acid (VPA), a histone deacetylase inhibitor, suppressed cell growth and upregulated the expression of Notch1 and SSTR2. A combination therapy with VPA and the SSTR2-targeting cytotoxic conjugate CPT-SST strongly led to greater suppression, as compared to each alone. Our findings thus provide us with a promising clinical opportunity for enhanced cancer therapy using combinations of Notch1-activating agents and SSTR2-targeting agents.


Subject(s)
Receptor, Notch1/physiology , Receptors, Somatostatin/physiology , Signal Transduction/physiology , Somatostatin/physiology , Uterine Cervical Neoplasms/prevention & control , Animals , Cell Cycle Checkpoints , Cell Proliferation , Colforsin/pharmacology , Cyclic AMP/metabolism , Female , HeLa Cells , Humans , Mice , Receptors, Somatostatin/antagonists & inhibitors , Uterine Cervical Neoplasms/pathology
10.
J Biol Chem ; 287(1): 382-392, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22084247

ABSTRACT

Janus kinase 1/signal transducers and activators of transcription 3 (JAK1/STAT3) pathway is one of the recognized oncogenic signaling pathways that frequently overactivated in a variety of human tumors. Despite rapid progress in elucidating the molecular mechanisms of activation of JAK/STAT pathway, the processes that regulate JAK/STAT deactivation need to be further clarified. Here we demonstrate that CUE domain-containing 2 (CUEDC2) inhibits cytokine-induced phosphorylation of JAK1 and STAT3 and the subsequent STAT3 transcriptional activity. Further analysis by a yeast two-hybrid assay showed that CUEDC2 could engage in a specific interaction with a key JAK/STAT inhibitor, SOCS3 (suppressors of cytokine signaling 3). The interaction between CUEDC2 and SOCS3 is required for the inhibitory effect of CUEDC2 on JAK1 and STAT3 activity. Additionally, we found CUEDC2 functions collaboratively with SOCS3 to inhibit JAK1/STAT3 signaling by increasing SOCS3 stability via enhancing its association with Elongin C. Therefore, our findings revealed a new biological activity for CUEDC2 as the regulator of JAK1/STAT3 signaling and paved the way to a better understanding of the mechanisms by which SOCS3 has been linked to suppression of the JAK/STAT pathway.


Subject(s)
Carrier Proteins/metabolism , Janus Kinase 1/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Elongin , Enzyme Activation , Humans , Phosphorylation , Protein Stability , Proteolysis , Suppressor of Cytokine Signaling 3 Protein , Transcription Factors/metabolism , Transcription, Genetic
11.
J Clin Invest ; 120(8): 2829-41, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628200

ABSTRACT

Activating mutations in Ras proteins are present in about 30% of human cancers. Despite tremendous progress in the study of Ras oncogenes, many aspects of the molecular mechanisms underlying Ras-induced tumorigenesis remain unknown. Through proteomics analysis, we previously found that the protein Gankyrin, a known oncoprotein in hepatocellular carcinoma, was upregulated during Ras-mediated transformation, although the functional consequences of this were not clear. Here we present evidence that Gankyrin plays an essential role in Ras-initiated tumorigenesis in mouse and human cells. We found that the increased Gankyrin present following Ras activation increased the interaction between the RhoA GTPase and its GDP dissociation inhibitor RhoGDI, which resulted in inhibition of the RhoA effector kinase Rho-associated coiled coil-containing protein kinase (ROCK). Importantly, Gankyrin-mediated ROCK inhibition led to prolonged Akt activation, a critical step in activated Ras-induced transformation and tumorigenesis. In addition, we found that Gankyrin is highly expressed in human lung cancers that have Ras mutations and that increased Gankyrin expression is required for the constitutive activation of Akt and tumorigenesis in these lung cancers. Our findings suggest that Gankyrin is a key regulator of Ras-mediated activation of Akt through inhibition of the downstream RhoA/ROCK pathway and thus plays an essential role in Ras-induced tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Genes, ras , Lung Neoplasms/etiology , Signal Transduction , Transcription Factors/physiology , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/physiology , Animals , Guanine Nucleotide Dissociation Inhibitors/physiology , Humans , Mice , NIH 3T3 Cells , PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins c-akt/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein
12.
J Biol Chem ; 285(19): 14356-65, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20231278

ABSTRACT

Nuclear factor-kappaB (NF-kappaB)-based signaling regulates diverse biological processes, and its deregulation is associated with various disorders including autoimmune diseases and cancer. Identification of novel factors that modulate NF-kappaB function is therefore of significant importance. The Mastermind-like 1 (MAML1) transcriptional co-activator regulates transcriptional activity in the Notch pathway and is emerging as a co-activator of other pathways. In this study, we found that MAML1 regulates NF-kappaB signaling via two mechanisms. First, MAML1 co-activates the NF-kappaB subunit RelA (p65) in NF-kappaB-dependent transcription. Second, MAML1 causes degradation of the inhibitor of NF-kappaB (IkappaBalpha). Maml1-deficient mouse embryonic fibroblasts showed impaired tumor necrosis factor-alpha (TNFalpha)-induced NF-kappaB responses. Moreover, MAML1 expression level directly influences cellular sensitivity to TNFalpha-induced cytotoxicity. In vivo, mice deficient in the Maml1 gene exhibited spontaneous cell death in the liver, with a large increase in the number of apoptotic hepatic cells. These findings indicate that MAML1 is a novel modulator for NF-kappaB signaling and regulates cellular survival.


Subject(s)
I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/physiology , Signal Transduction , Transcription Factor RelA/metabolism , Transcription Factors/physiology , Animals , Apoptosis , Blotting, Western , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival , Cells, Cultured , Electrophoretic Mobility Shift Assay , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Immunoprecipitation , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
13.
J Proteome Res ; 6(11): 4397-406, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17894482

ABSTRACT

A global understanding of ubiquitinated proteins in vivo is key to unraveling the biological significance of ubiquitination. There are, however, a few effective screening methods for rapid analysis of ubiquitinated proteins. In the current study, we designed a cell-based cDNA expression array combined with cell imaging for the rapid identification of polyubiquitinated proteins, which normally accumulate to form the unique "dot" structure following inhibition of ubiquitin proteasomes. The array consisted of 112 cDNAs encoding key components of major cellular pathways and potential targets of polyubiquitination. Among them, 40 proteins formed accumulation dots in response to proteasome inhibitor, MG-132, treatment. More importantly, 24 of those 40 proteins, such as MAPKAPK3, NLK, and RhoGDI2, are previously not known as the targets of ubiquitin. We further validated our findings by examining the endogenous counterparts of some of these proteins and found that those endogenous proteins form a similar "dot" structure. Immunoprecipitation assays confirmed that these accumulated proteins are polyubiquitinated. Our results demonstrate that this large-scale application of cell-based arrays represents a novel global approach in identifying candidates of the polyubiquitinated proteins. Therefore, the technique utilized here will facilitate future research on ubiquitination-regulated cell signaling.


Subject(s)
Proteins/chemistry , Proteomics/methods , Ubiquitin/chemistry , Cell Line, Tumor , DNA, Complementary/metabolism , Electrophoresis, Gel, Two-Dimensional , Guanine Nucleotide Dissociation Inhibitors/metabolism , HeLa Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Leupeptins/pharmacology , Proteasome Inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
14.
EMBO J ; 26(7): 1831-42, 2007 Apr 04.
Article in English | MEDLINE | ID: mdl-17347654

ABSTRACT

Accumulated evidence indicates that progesterone receptors (PR) are involved in proliferation of breast cancer cells and are implicated in the development of breast cancer. In this paper, a yeast two-hybrid screen for PR led to the identification of CUE domain containing 2 (CUEDC2), whose function is unknown. Our results demonstrate that CUEDC2 interacts with PR and promotes progesterone-induced PR degradation by the ubiquitin-proteasome pathway. The inhibition of endogenous CUEDC2 by siRNA nearly abrogated the progesterone-induced degradation of PR, suggesting that CUEDC2 is involved in progesterone-induced PR ubiquitination and degradation. Moreover, we identify the sumoylation site Lys-388 of PR as the target of CUEDC2-promoted ubiquitination. CUEDC2 decreases the sumoylation while promoting ubiquitination on Lys-388 of PRB. We also show that CUEDC2 represses PR transactivation, inhibits the ability of PR to stimulate rapid MAPK activity, and impairs the effect of progesterone on breast cancer cell growth. Therefore, our results identify a key post-translational mechanism that controls PR protein levels and for the first time provide an important insight into the function of CUEDC2 in breast cancer proliferation.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Receptors, Progesterone/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Ligands , Membrane Proteins/genetics , Mutant Proteins/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Protein Interaction Mapping , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary/drug effects , Receptors, Progesterone/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
15.
J Proteome Res ; 5(10): 2815-23, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17022653

ABSTRACT

We implemented a proteomics approach to the systematical analysis of the alterations in the proteome of NIH3T3 cells transformed by oncogenic H-RasV12. Forty-four proteins associated with Ras-mediated transformation have been identified, and 28 proteins were not previously reported. RT-PCR analysis showed that approximately 44% of target proteins identified showed concomitant changes in mRNA abundance. A principal finding was the up-regulation of gankyrin, which was the first evidence to show that gankyrin pathway was implicated in Ras-activated transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Oncogene Protein p21(ras)/genetics , Proteome/analysis , Proteomics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Humans , Mice , NIH 3T3 Cells , Proteome/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
16.
Nucleic Acids Res ; 34(19): 5552-66, 2006.
Article in English | MEDLINE | ID: mdl-17020914

ABSTRACT

Progesterone receptor (PR) plays a critical role in cell proliferation and differentiation, and its transcriptional activity is known to be modulated by cofactor proteins. In the present study, we demonstrated that in the presence of progesterone, protein inhibitor of activated STAT-3 (PIAS3) significantly inhibited the PR transcriptional activity and the expression of progesterone-responsive genes. Reduction of endogenous PIAS3 by PIAS3 small-interfering RNA enhanced PR transactivation in a ligand-dependent manner. PIAS3 interacted with PR both in vitro and in vivo and the interaction was enhanced by progesterone. Furthermore, our findings suggested that PIAS3 strongly induced PRB sumoylation at three sites, Lys-7, Lys-388 and Lys-531. In addition, novel roles in PRB nuclear retention and transactivation were identified for these sites. Our data also suggested that PIAS3 was recruited in a largely hormone-dependent manner in response to a progesterone-responsive promoter. Finally, we demonstrated that PIAS3 inhibited the DNA-binding activity of PR and influenced its nuclear export as well as PR transactivation. Taken together, these data strongly suggested that PIAS3 played an important physiological role in PR function.


Subject(s)
Cell Nucleus/chemistry , Molecular Chaperones/metabolism , Protein Inhibitors of Activated STAT/metabolism , Receptors, Progesterone/metabolism , Transcriptional Activation , Animals , Humans , Progesterone/antagonists & inhibitors , Promoter Regions, Genetic , Protein Processing, Post-Translational , Receptors, Progesterone/analysis , Receptors, Progesterone/antagonists & inhibitors , Small Ubiquitin-Related Modifier Proteins/metabolism
17.
Biochem Biophys Res Commun ; 345(1): 355-61, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16678796

ABSTRACT

Recent reports have shown that MDM2 may attenuate hypertrophy of cardiac myocytes. However, mechanism of MDM2 involving in this process is unclear. In this study, we identified a novel specific MDM2-binding protein TCAP by the yeast two-hybrid screen. It was validated by GST pull-down and co-immunoprecipitation assays. Confocal analysis showed that MDM2 and TCAP co-localized in the nucleus, and elevated MDM2 expression could alter the subcellular localization of TCAP. Notably, MDM2 downregulated the protein level of TCAP through the proteasomal pathway, and this downregulation was inhibited by p14(ARF). In addition, our results suggested that the degradation of TCAP by MDM2 was through the ubiquitin-independent pathway. Given that TCAP is a key component involving in the cardiac hypertrophy, the degradation of TCAP by MDM2 might be connected with the roles of MDM2 in cardiac hypertrophy. Further investigation will focus on the biological significance of MDM2-TCAP interaction in cardiac hypertrophy.


Subject(s)
Muscle Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Sarcomeres/metabolism , Subcellular Fractions/metabolism , Connectin , Down-Regulation , Muscle Proteins/chemistry , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins c-mdm2/chemistry , Sarcomeres/chemistry , Subcellular Fractions/chemistry
18.
Biochem Biophys Res Commun ; 345(2): 660-8, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16696944

ABSTRACT

Stat3 plays important roles in the development of breast malignancies and oncogenesis. In the present study, a palindromic cis-acting element displaying repression activity in breast cancer cells expressing low level of Her2 was found in Her2 promoter. Deletion analysis showed that the novel element was located within Pal2 region spanning nucleotides -529 to -505. The sequence analysis of Pal2 region revealed a DNA sequence (TTAAGATAA) homologous to the binding site of Stat3, starting from position -529 to -521bp. By reporter assay, Pal2 was found to be regulated by constitutive activated Stat3C. A stimulatory effect both on Her2 mRNA and protein expressions was observed in MCF-7 cells stably expressing Stat3C, suggesting that Stat3 regulated Her2 expression. Using ChIP assays the binding of Stat3 to Her2 promoter was confirmed. The data obtained in this study indicate constitutive activated Stat3 regulates Her2 expression. Further investigation of differential effects of Stat3 exerting on breast cancer cells expressing Her2 at different levels will provide more insights into the roles of Stat3 in Her2 expression as well as the regulation of diverse biological activities.


Subject(s)
Genes, erbB-2/genetics , Promoter Regions, Genetic , STAT3 Transcription Factor/physiology , Base Sequence , Binding Sites , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Genes, erbB-2/physiology , Humans , Microchip Analytical Procedures , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
19.
Biochem Biophys Res Commun ; 344(3): 727-34, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16631117

ABSTRACT

SOX4 is a member of SOX transcriptional factor family that is crucial for many cellular processes. In this study, a yeast two-hybrid screening of human mammary cDNA library identified human ubiquitin-conjugating enzyme 9 (hUbc9) that interacted with SOX4. This interaction was confirmed by GST pull-down in vitro and co-immunoprecipitation assays in vivo. Deletion mapping demonstrated that HMG-box domain of SOX4 is required to mediate the interaction with Ubc9 in yeast. Furthermore, confocal microscopy showed that Ubc9 co-localized with SOX4 in the nucleus. Luciferase assays found that Ubc9 specifically repressed SOX4 transcriptional activity in 293T cells. We further demonstrated that Ubc9 could functionally repress the transcriptional activity of endogenous SOX4 induced by progesterone in T47D cells. The C93S mutant of Ubc9, which abrogates SUMO-1 conjugation activity, did not abolish the ability to repress SOX4 activity. It shows that Ubc9 interacts with SOX4 and represses its transcriptional activity independent of its SUMO-1-conjugating activity.


Subject(s)
High Mobility Group Proteins/metabolism , Suppression, Genetic/physiology , Trans-Activators/metabolism , Transcription, Genetic/physiology , Transcriptional Activation/physiology , Ubiquitin-Conjugating Enzymes/metabolism , High Mobility Group Proteins/genetics , Humans , Protein Binding , SOXC Transcription Factors , Trans-Activators/genetics , Ubiquitin-Conjugating Enzymes/genetics
20.
Electrophoresis ; 26(1): 268-79, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15624164

ABSTRACT

Two-dimensional electrophoresis (2-DE) was used to profile the proteins of leukemic cells from 61 cases of akute leukemia (AL) characterized by the French-American-British (FAB) classification. The differentially expressed protein spots were identified by matrix assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS) and electrospray ionization-tandem MS (ESI-MS/MS). The distinct protein profiles (DPPs) of AL FAB subtypes were explored successfully, including acute myeloid leukemia (AML), its subtypes (M2, M3, and M5), and acute lymphoid leukemia (ALL), which were homogeneous within different samples of the same subgroup but clearly differed from all other subgroups. We also found a group of proteins differentially expressed between AL cells and normal white blood cells. Among the DPPs of AL subtypes, some proteins have been reported, but most of them were first reported here to mark AML differentiation and to discriminate AML from ALL. These data show that 2-DE protein profiling could be used as an analytical tool for facilitating molecular definition of human AL classification and understanding the mechanism of leukemogensis, and the extension of the present analysis to the currently less well-defined AL will identify additional subgroups and may promote the identification of new targets for specific treatment approaches.


Subject(s)
Electrophoresis, Gel, Two-Dimensional , Leukemia/classification , Neoplasm Proteins/analysis , Acute Disease , Adolescent , Adult , Biomarkers, Tumor/analysis , Female , Humans , Male , Peptide Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...