Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Differ ; 25(12): 2195-2208, 2018 12.
Article in English | MEDLINE | ID: mdl-29795334

ABSTRACT

TP53 mutations occur in many different types of cancers that produce mutant p53 proteins. The mutant p53 proteins have lost wild-type p53 activity and gained new functions that contribute to malignant tumor progression. Different p53 mutations create distinct profiles in loss of wild-type p53 activity and gain of functions. Targeting the consequences generated by the great number of p53 mutations would be extremely complex. Therefore, in this study we used a workaround and took advantage of the fact that mutant p53 cannot bind H2AX. Using this, we developed a new approach to repress the acquisition of mutant p53 functions. We show here that the delivery of a circular RNA circ-Ccnb1 inhibited the function of three p53 mutations. By microarray analysis and real-time PCR, we detected decreased circ-Ccnb1 expression levels in patients bearing breast carcinoma. Ectopic delivery of circ-Ccnb1 inhibited tumor growth and extended mouse viability. Using proteomics, we found that circ-Ccnb1 precipitated p53 in p53 wild-type cells, but instead precipitated Bclaf1 in p53 mutant cells. Further experiments showed that H2AX serves as a bridge, linking the interaction of circ-Ccnb1 and wild-type p53, thus allowing Bclaf1 to bind Bcl2 resulting in cell survival. In the p53 mutant cells, circ-Ccnb1 formed a complex with H2AX and Bclaf1, resulting in the induction of cell death. We found that this occurred in three p53 mutations. These results shed light on the possible development of new approaches to inhibit the malignancy of p53 mutations.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Nucleic Acid Conformation , RNA/chemistry , RNA/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Animals , Binding Sites/drug effects , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Female , HEK293 Cells , Humans , Injections, Intraperitoneal , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Molecular Docking Simulation , Mutation , Proteomics , RNA/genetics , Tumor Suppressor Protein p53/metabolism
2.
Mol Cancer Res ; 11(5): 443-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23449934

ABSTRACT

Versican is highly expressed during the early stages of tissue development and its expression is elevated during wound repair and tumor growth. There is little literature on the potential role of breast cancer stem cells on the cellular-extracellular matrix interactions involving versican. An anti-versican short hairpin RNA (shRNA) was used to observe the effect of reduction of versican on breast cancer self-renewal. A versican G3 construct was exogenously expressed in breast cancer cell lines. Colony formation and mammosphere formation assays were conducted; flow cytometry was applied to analyze the prevalence of side population cells. The versican G3- and vector-transfected 66c14 cells were injected transdermally into BALB/c mice as a 10-fold dilution series from 1 × 10(5) to 1 × 10(2) cells per mouse. Versican G3 domain enhanced breast cancer self-renewal in both experimental in vitro and in vivo models. Versican G3-transfected cells contained high levels of side population cells, formed more mammospheres when cultured in the serum-free medium, and formed a greater number and larger colonies. Reduction of versican's functionality through anti-versican shRNA or knocking out the EGF-like motifs reduced the effect of versican on enhancing mammosphere and colony formation. Versican-enhanced self-renewal played a role in enhanced chemotherapeutic drug resistance, relating partly to the upregulated expression of EGF receptor (EGFR) signaling. Versican is highly expressed in breast cancer progenitor cells and was maintained at high levels before cell differentiation. Overexpression of versican enhanced breast cancer self-renewal through EGFR/AKT/GSK-3ß (S9P) signaling and conferred resistant to chemotherapeutic drugs tested.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Versicans/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Signal Transduction , Transfection , Versicans/genetics
3.
PLoS One ; 6(11): e26396, 2011.
Article in English | MEDLINE | ID: mdl-22096483

ABSTRACT

Overexpression of EGFR and versican has been reported in association with breast cancers. Considered oncogenic, these molecules may be attractive therapeutic targets. Possessing anti-apoptotic and drug resistant properties, overexpression of these molecules is accompanied by selective sensitization to the process of apoptosis. In this study, we exogenously expressed a versican G3 construct in breast cancer cell lines and analyzed the effects of G3 on cell viability in fetal bovine serum free conditioned media and evaluated the effects of apoptotic agent C2-ceramide, and chemotherapeutic agents including Docetaxel, Doxorubicin, and Epirubicin. Versican G3 domain enhanced tumor cell resistance to apoptosis when cultured in serum free medium, Doxorubicin, or Epirubicin by up-regulating pERK and GSK-3ß (S9P). However, it could be prevented by selective EGFR inhibitor AG 1478 and selective MEK inhibitor PD 98059. Both AG 1478 and PD 98059 enhanced expression of pSAPK/JNK, while selective JNK inhibitor SP 600125 enhanced expression of GSK-3ß (S9P). Versican G3 promoted cell apoptosis induced by C2-ceramide or Docetaxel by enhancing expression of pSAPK/JNK and decreasing expression of GSK-3ß (S9P), an observation blocked by AG 1478 or SP 6000125. Inhibition of endogenous versican expression by siRNA or reduction of versican G3's expression by linking G3 with 3'UTR prevented G3 modulated cell apoptosis. The dual roles of G3 in modulating breast cancer cell resistance to chemotherapeutic agents may in part explain a potential mechanism for breast cancer cell resistance to chemotherapy and EGFR therapy. The apoptotic effects of chemotherapeutics depend upon the activation and balance of down stream signals in the EGFR pathway. GSK-3ß (S9P) appears to function as a key checkpoint in this balance of apoptosis and anti-apoptosis. Investigation and potential consideration of targeting GSK-3ß (S9P) merits further study.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/metabolism , Green Fluorescent Proteins/metabolism , Versicans/metabolism , Animals , Anthracenes/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/drug effects , Cell Survival/genetics , Docetaxel , Doxorubicin/pharmacology , Epirubicin/pharmacology , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/antagonists & inhibitors , Humans , Mice , Quinazolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Taxoids/pharmacology , Tyrphostins/pharmacology , Versicans/genetics
4.
PLoS One ; 5(11): e13828, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21079779

ABSTRACT

Increased versican expression in breast tumors is predictive of relapse and has negative impact on survival rates. The C-terminal G3 domain of versican influences local and systemic tumor invasiveness in pre-clinical murine models. However, the mechanism(s) by which G3 influences breast tumor growth and metastasis is not well characterized. Here we evaluated the expression of versican in mouse mammary tumor cell lines observing that 4T1 cells expressed highest levels while 66c14 cells expressed low levels. We exogenously expressed a G3 construct in 66c14 cells and analyzed its effects on cell proliferation, migration, cell cycle progression, and EGFR signaling. Experiments in a syngeneic orthotopic animal model demonstrated that G3 promoted tumor growth and systemic metastasis in vivo. Activation of pERK correlated with high levels of G3 expression. In vitro, G3 enhanced breast cancer cell proliferation and migration by up-regulating EGFR signaling, and enhanced cell motility through chemotactic mechanisms to bone stromal cells, which was prevented by inhibitor AG 1478. G3 expressing cells demonstrated increased CDK2 and GSK-3ß (S9P) expression, which were related to cell growth. The activity of G3 on mouse mammary tumor cell growth, migration and its effect on spontaneous metastasis to bone in an orthotopic model was modulated by up-regulating the EGFR-mediated signaling pathway. Taken together, EGFR-signaling appears to be an important pathway in versican G3-mediated breast cancer tumor invasiveness and metastasis.


Subject(s)
Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/pathology , Signal Transduction , Versicans/physiology , Animals , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Transplantation , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Serine/metabolism , Transfection , Versicans/genetics , Versicans/metabolism
5.
Breast Cancer Res ; 9(4): R47, 2007.
Article in English | MEDLINE | ID: mdl-17662123

ABSTRACT

INTRODUCTION: Increased versican expression has been associated with local breast cancer invasiveness and a more aggressive tumor phenotype. The cellular mechanisms are not fully understood and this study evaluated versican G3 domain with its EGF-like motifs in influencing tumor invasion and metastasis. METHODS: One recombinant construct was synthesized (a signal peptide for product secretion and the versican G3 domain). The construct was stably transfected into human breast carcinoma MT-1 cells. Cell viability in vitro was evaluated in low serum and serum starvation conditions. In vivo study of tumor growth was evaluated in a nude mouse model. G3 effects on rodent vascular endothelial cells were evaluated in vitro on cell survival, apoptosis, migration, and vascular formation. The effects of VEGF, fibronectin, and G3 on vascular formation were examined. An intracardiac injection model of metastatic human breast carcinoma tested the effect of G3 on distant bony and soft tissue metastasis. Analysis of metastatic burden included histology, radiographs, and micro-CT quantification of osteolysis. RESULTS: A greater viability of cancer cells was observed in low serum and serum-free conditions in the presence of versican G3. Larger subcutaneous tumors were obtained in the G3 group following tumor cell injection into CD1 mice. G3 induced a greater degree of rodent vascular endothelial cell proliferation and migration in vitro. Simultaneous presence of fibronectin, VEGF, and G3 promoted endothelial cell migration in wound-healing assays as compared to the treatments containing none, one or two of these molecules. Systemic tumor burden to distant bony and soft tissue metastatic sites was greater in the G3 group using the intracardiac injection metastatic model. CONCLUSION: Versican G3 domain appears to be important in local and systemic tumor invasiveness of human breast cancer. Effects include enhancing cell viability, proliferation, migration and enhancing local tumor growth. Potential effects on angiogenesis include enhancing vascular endothelial proliferation, migration, and vessel formation. The interactions between tumor cells, surrounding stromal components and neo-vascularization in breast cancer may include interactions with VEGF and fibronectin. The propensity of versican G3 to influence tumor invasion to bone and the mechanisms of G3 mediated osteolysis warrants ongoing studies.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Versicans/pharmacology , Animals , Bone Neoplasms/metabolism , Breast Neoplasms/blood supply , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Chickens , Endothelium, Vascular/drug effects , Female , Fibronectins/metabolism , Fibronectins/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Osteolysis , Protein Structure, Tertiary , Transfection , Vascular Endothelial Growth Factor A/pharmacology
6.
J Cell Sci ; 117(Pt 24): 5887-95, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15522894

ABSTRACT

P-selectin glycoprotein ligand-1 (PSGL-1), a glycoprotein expressed on the cell surface of leukocytes, binds to selectins and mediates leukocyte rolling on the vascular endothelium. Here we report that PSGL-1 binds to the C-terminal (G3 domain) of the extracellular proteoglycan PG-M/versican. Cells transfected with PSGL-1 or a shorter form containing the binding site, or cells expressing endogenous PSGL-1 aggregate in the presence of versican or G3 product. The aggregation appears to be induced by G3 multimers that bind to PSGL-1 and form a network. Endogenous versican and/or G3-containing fragments also bind to PSGL-1 in human plasma. Removal of the endogenous G3-containing fragments reduces the effect of plasma on leukocyte aggregation. Finally, the roles of G3-containing fragments in leukocyte aggregation were confirmed in a mouse model. Taken together, our results strongly support a physiologically relevant role for PSGL-1/versican binding and may have implications in the immunoresponse.


Subject(s)
Chondroitin Sulfate Proteoglycans/physiology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/physiology , Amino Acid Motifs , Binding Sites , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , DNA Primers/chemistry , DNA, Complementary/metabolism , Disulfides/chemistry , Dithiothreitol/pharmacology , Gene Library , Glutathione Transferase/metabolism , Glycoproteins/chemistry , Humans , Leukocytes/cytology , Leukocytes/metabolism , Matrix Metalloproteinases/chemistry , Models, Biological , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry , Signal Transduction , Transfection , Two-Hybrid System Techniques , Versicans
7.
Life Sci ; 73(26): 3399-413, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14572881

ABSTRACT

The functional role of versican in influencing intervertebral disc cell adhesion and proliferation was analyzed in bovine intervertebral disc. We have previously demonstrated the C-terminal globular G3 (or selectin-like) domain of versican to influence mesenchymal chondrogenesis and fibroblast proliferation in vitro. For this study, a versican G3 expression construct was generated to examine the role of the G3 domain of versican. Nucleus pulposus and annulus fibrosus cells were isolated from adult bovine caudal discs using sequential enzymatic digestion and versican expression characterized by RT-PCR. In cell proliferation assays, we observed that there was greater cellular proliferation in the presence of versican G3 for both disc cell types. The higher proliferation rate of annulus fibrosus cells when compared to nucleus pulposus cells seeded in monolayer supports heterogeneity of intervertebral disc cell populations. The presence of versican G3 construct enhanced the adhesion of isolated nucleus pulposus and annulus fibrosus cells approximately 4 to 6 fold, respectively. Cellular adhesion was greater in the presence of versican G3 in a dose dependent manner. G3 product was purified using affinity columns, and the purified G3 also enhanced cell adhesion.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Intervertebral Disc/metabolism , Proteoglycans/metabolism , Animals , Cattle , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/genetics , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Gene Expression , In Vitro Techniques , Intervertebral Disc/cytology , Intervertebral Disc/drug effects , Lectins, C-Type , Protein Structure, Tertiary , Proteoglycans/chemistry , Proteoglycans/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Versicans
8.
Biochemistry ; 42(27): 8332-41, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12846582

ABSTRACT

The extracellular matrix plays a critical role in maintaining tissue integrity. Among the matrix molecules, the large aggregating chondroitin sulfate proteoglycans are the major structural molecules and are the primary contributors to the stability for some tissues such as cartilage. The notable exceptions are nanomelic cartilage and arthritic cartilage: the former contains a point mutation leading to a stop codon before translating to the C-terminal G3 domain; the latter contains a large proportion of aggrecan from which the G3 domain has been cleaved. These phenomena suggest that the G3 domain may be important in cartilage stability. Here, we demonstrated for the first time that the G3 domains of aggrecan and another proteoglycan, PG-M/versican, formed intermolecular disulfide bonds, and all subdomains were involved. Further studies indicated that each of the 10 cysteine residues of the aggrecan G3 domain could potentially form intermolecular disulfide bonds in vitro. The disulfide bonds were disrupted in the presence of reducing reagent beta-mercaptoethanol and dithiothreitol. As a result, normal chondrocyte-matrix interaction was disrupted, and the structure of the extracellular matrix was altered. Furthermore, disruption of disulfide bonds also reduced the role of PG-M/versican G3 domain in mediating cell adhesion. Our study provides strong evidence of the importance of proteoglycan interactions through intermolecular disulfide bonds in cartilage firmness and cell-matrix stability.


Subject(s)
Disulfides/chemistry , Extracellular Matrix Proteins , Proteoglycans/chemistry , Aggrecans , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Culture Media , DNA Primers , Lectins, C-Type , Models, Molecular , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...