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.
BMC Cancer ; 16: 575, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27484185

ABSTRACT

BACKGROUND: Active fibroblasts, the predominant and the most active cells of breast cancer stroma, are responsible for tumor growth and spread. However, the molecular mediators and pathways responsible for stromal fibroblast activation, and their paracrine pro-carcinogenic effects are still not well defined. The CHEK2 tumor suppressor gene codes for a protein kinase, which plays important roles in the cellular response to various genotoxic stresses. METHODS: Immunoblotting, quantitative RT-PCR and Immunofluorescence were used to assess the expression of CHEK2 in different primary breast fibroblasts and in tissues. The effect of CHEK2 on the expression and secretion of SDF-1 and IL-6 was evaluated by immunoblotting and ELISA. The WST-1 colorimetric assay was used to assess cell proliferation, while the BD BioCoat Matrigel invasion chambers were utilized to determine the effects of CHEK2 on the migratory and the invasiveness capacities of breast stromal fibroblasts as well as breast cancer cells. RESULTS: We have shown that CHEK2 is down-regulated in most cancer-associated fibroblasts (CAFs) as compared to their corresponding tumor counterpart fibroblasts (TCFs) at both the mRNA and protein levels. Interestingly, CHEK2 down-regulation using specific siRNA increased the expression/secretion of both cancer-promoting cytokines SDF-1 and IL-6, and transdifferentiated stromal fibroblasts to myofibroblasts. These cells were able to enhance the proliferation of non-cancerous epithelial cells, and also boosted the migration/invasion abilities of breast cancer cells in a paracrine manner. The later effect was SDF-1/IL-6-dependent. Importantly, ectopic expression of CHEK2 in active CAFs converted these cells to a normal state, with lower migration/invasion capacities and reduced paracrine pro-carcinogenic effects. CONCLUSION: These results indicate that CHEK2 possesses non-cell-autonomous tumor suppressor functions, and present the Chk2 protein as an important mediator in the functional interplay between breast carcinomas and their stromal fibroblasts.


Subject(s)
Breast Neoplasms/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Chemokine CXCL12/metabolism , Interleukin-6/metabolism , Stromal Cells/cytology , Breast/cytology , Breast/metabolism , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CXCL12/genetics , Coculture Techniques , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/genetics , Paracrine Communication , Stromal Cells/metabolism
2.
J Pathol ; 231(4): 457-65, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24009142

ABSTRACT

There is evidence that normal breast stromal fibroblasts (NBFs) suppress tumour growth, while cancer-associated fibroblasts (CAFs) promote tumourigenesis through functional interactions with tumour cells. Little is known about the biology and the carcinogenic potential of stromal fibroblasts present in histologically normal surgical margins (TCFs). Therefore, we first undertook gene expression analysis on five CAF/TCF pairs from breast cancer patients and three NBF samples (derived from mammoplasties). This comparative analysis revealed variation in gene expression between these three categories of cells, with a TCF-specific gene expression profile. This variability was higher in TCFs than in their paired CAFs and also NBFs. Cytokine arrays show that TCFs have a specific secretory cytokine profile. In addition, stromal fibroblasts from surgical margins expressed high levels of α-SMA and SDF-1 and exhibited higher migratory/invasiveness abilities. Indirect co-culture showed that TCF cells enhance the proliferation of non-cancerous mammary epithelial cells and the epithelial-to-mesenchymal transition of breast cancer cells. Moreover, TCF and CAF cells increased the level of PCNA, MMP-2 and the phosphorylated/activated form of Akt in normal breast luminal fibroblasts in a paracrine manner. Furthermore, TCFs were able to promote the formation and growth of humanized orthotopic breast tumours in nude mice. Interestingly, these TCF phenotypes and the extent of their effects were intermediate between those of NBFs and CAFs. Together, these results indicate that stromal fibroblasts located in non-cancerous tissues exhibit a tumour-promoting phenotype, indicating that their presence post-surgery may play important roles in cancer recurrence.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Fibroblasts/physiology , Animals , Breast Neoplasms/surgery , Cell Movement/physiology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Chemokine CXCL12/biosynthesis , Cytokines/biosynthesis , Epithelial-Mesenchymal Transition , Female , Fibroblasts/metabolism , Gene Expression Profiling/methods , Heterografts , Humans , Interleukin-6/biosynthesis , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis/methods , Stromal Cells/metabolism , Stromal Cells/physiology , Vascular Endothelial Growth Factor A/biosynthesis
3.
Mol Carcinog ; 51(12): 930-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-21976111

ABSTRACT

The universal cyclin-dependent kinase inhibitor p21(WAF1/Cip1) promotes cell cycle arrest and inhibits apoptosis in response to UV-induced DNA damage. Since the protein kinase ATR plays a major role in the cellular response to these carcinogenic lesions, we investigated the possible role of ATR in the modulation of p21(WAF1/Cip1) expression in response to UVC radiation. We have shown that p21(WAF1/Cip1) is up-regulated in human fibroblast and epithelial cells, but only in response to low UV fluences and low passage cells. Importantly, this up-regulation is ATR-dependent. In fact, in ATR-deficient or caffeine-treated cells UV light rather down-regulated the p21(WAF1/Cip1) protein through SKP2-dependent ubiquitination and degradation via the proteasomal pathway. Furthermore, we present evidence that ATR inhibits apoptosis in response to low fluences of UV light, through inhibiting the cleavage of caspase 3 and PARP as well as the repression of the proapoptotic proteins BAX and BAK. Interestingly, ATR is also required for the stability of the p21(WAF1/Cip1) protein in absence of genotoxic stress. Together, these results indicate that during the cellular response to low UVC fluences the ATR protein kinase up-regulates p21(WAF1/Cip1) and inhibits apoptosis. © 2011 Wiley Periodicals, Inc.


Subject(s)
Apoptosis/radiation effects , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Protein Serine-Threonine Kinases/physiology , Ultraviolet Rays , Up-Regulation/radiation effects , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle , Dose-Response Relationship, Radiation , Electrophoresis, Polyacrylamide Gel , Humans , Ubiquitination , Up-Regulation/physiology
4.
PLoS One ; 6(7): e21111, 2011.
Article in English | MEDLINE | ID: mdl-21799732

ABSTRACT

BACKGROUND: The cyclin-D/CDK4,6/p16(INK4a)/pRB/E2F pathway, a key regulator of the critical G1 to S phase transition of the cell cycle, is universally disrupted in human cancer. However, the precise function of the different members of this pathway and their functional interplay are still not well defined. METHODOLOGY/PRINCIPAL FINDINGS: We have shown here that the tumor suppressor p16(INK4a) protein positively controls the expression of cyclin D1 and E2F1 in both human and mouse cells. p16(INK4a) stabilizes the mRNAs of the corresponding genes through negative regulation of the mRNA decay-promoting AUF1 protein. Immunoprecipitation of AUF1-associated RNAs followed by RT-PCR indicated that endogenous AUF1 binds to the cyclin D1 and E2F1 mRNAs. Furthermore, AUF1 down-regulation increased the expression levels of these genes, while concurrent silencing of AUF1 and p16(INK4a), using specific siRNAs, restored normal expression of both cyclinD1 and E2F1. Besides, we have shown the presence of functional AU-rich elements in the E2F1 3'UTR, which contributed to p16/AUF1-mediated regulation of E2F1 post-transcriptional events in vivo. Importantly, genome-wide gene expression microarray analysis revealed the presence of a large number of genes differentially expressed in a p16(INK4a) -dependent manner, and several of these genes are also members of the AUF1 and E2F1 regulons. We also present evidence that E2F1 mediates p16-dependent regulation of several pro- and anti-apoptotic proteins, and the consequent induction of spontaneous as well as doxorubicin-induced apoptosis. CONCLUSION/SIGNIFICANCE: These findings show that the cyclin-dependent kinase inhibitor p16( INK4a) is also a modulator of transcription and apoptosis through controlling the expression of two major transcription regulators, AUF1 and E2F1.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , E2F1 Transcription Factor/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , 3' Untranslated Regions/genetics , Animals , Apoptosis/drug effects , Base Sequence , Cell Line , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Doxorubicin/pharmacology , E2F1 Transcription Factor/genetics , GC Rich Sequence/genetics , Gene Expression Regulation/drug effects , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Mol Cancer Res ; 9(3): 311-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21270107

ABSTRACT

The tumor suppressor p16(INK4A), a phosphoprotein that exists in human cells under both phosphorylated and nonphosphorylated forms, plays crucial roles during the cellular response to UV light. However, it is still unclear how this protein is activated in response to this carcinogenic agent. We have shown here that UVC upregulates p16(INK4A) and the phosphorylated form of the protein at the 4 serine sites; Ser-7, Ser-8, Ser-140, and Ser-152. This accumulation of p16(INK4A) occurred through increasing the stability of both forms of the protein. Importantly, phospho-p16(INK4A) showed much higher stability, and UV treatment strongly increased its level in absence of de novo protein synthesis. Furthermore, we have shown that the UV-dependent upregulation of both forms of p16(INK4A) is under the control of the protein kinase Atr, which suppresses their UVC-dependent proteasomal degradation. Interestingly, although this degradation is ubiquitin-related for p16(INK4A) through the Skp2 ubiquitin ligase protein, it is ubiquitin-independent for the phosphorylated form. In addition, we present clear evidence that Skp2 is upregulated in ATR-deficient cells, leading to the downregulation of the p27(Kip1) protein in response to UVC light. Moreover, we have shown a preferential association of endogeneous phospho-p16(INK4A) with Cdk4. This association increased following UV-treatment mainly for p16(INK4A) phosphorylated at Ser-140 and Ser-152. Besides, we have shown that Atr regulates UV-related p16/Cdk4-dependent and -independent phosphorylation of pRB and G1 cell cycle delay. Together, these results indicate that p16(INK4A) and p27(Kip1) are key targets in the Atr-dependent signaling pathway in response to UV damage.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Serine-Threonine Kinases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ultraviolet Rays , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Line , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , S-Phase Kinase-Associated Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitination/genetics , Ultraviolet Rays/adverse effects , Up-Regulation/genetics
6.
Breast Cancer Res ; 12(4): R48, 2010.
Article in English | MEDLINE | ID: mdl-20626886

ABSTRACT

INTRODUCTION: B7-H1 (PD-L1, CD274) is a T cell inhibitory molecule expressed in many types of cancer, leading to immune escape of tumor cells. Indeed, in previous reports we have shown an association of B7-H1 expression with high-risk breast cancer patients. METHODS: In the current study, we used immunohistochemistry, immunofluorescence and Western blot techniques to investigate the effect of neoadjuvant chemotherapy on the expression of B7-H1 in breast cancer cells. RESULTS: Among tested chemotherapeutic agents, doxorubicin was the most effective in downregulating cell surface expression of B7-H1 in vitro. These results were validated in vivo in a xenograft mouse model, as well as in murine heart tissue known to constitutively express B7-H1. The doxorubicin-dependent cell surface downregulation of B7-H1 was accompanied by an upregulation of B7-H1 in the nucleus. This re-distribution of B7-H1 was concurrent with a similar translocation of phosphorylated AKT to the nucleus. Inhibition of the PI3K/AKT pathway abrogated the doxorubicin-mediated nuclear up-regulation of B7-H1, suggesting an involvement of PI3K/AKT pathway in the nuclear up-regulation of B7-H1. Interestingly, siRNA knock down of B7-H1 lead to an increase in spontaneous apoptosis, as well as doxorubicin-induced apoptosis, which indicates an anti-apoptotic role for B7-H1 in breast cancer cells. The novel discovery of B7-H1 expression in the nuclei of breast cancer cells suggests that B7-H1 has functions other than inhibition of T cells. CONCLUSIONS: Our findings explain the previously reported immunomodulatory effect of anthracyclines on cancer cells, and provide a link between immunoresistance and chemoresistance. Finally these results suggest the use of dual combinatorial agents to inhibit B7-H1 beside chemotherapy, in breast cancer patients.


Subject(s)
Antigens, CD/metabolism , Apoptosis/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antigens, CD/genetics , B7-H1 Antigen , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Down-Regulation/drug effects , Female , Humans , Immunohistochemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
7.
Mol Carcinog ; 49(3): 302-14, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20025076

ABSTRACT

Medulloblastoma is an aggressive primary brain tumor that arises in the cerebellum of children and young adults. The Sonic Hedgehog (Shh) signaling pathway that plays important roles in the pathology of this aggressive disease is a promising therapeutic target. In the present report we have shown that curcumin has cytotoxic effects on medulloblastoma cells. Curcumin suppressed also cell proliferation and triggered cell-cycle arrest at G(2)/M phase. Moreover, curcumin inhibited the Shh-Gli1 signaling pathway by downregulating the Shh protein and its most important downstream targets GLI1 and PTCH1. Furthermore, curcumin reduced the levels of beta-catenin, the activate/phosphorylated form of Akt and NF-kappaB, which led to downregulating the three common key effectors, namely C-myc, N-myc, and Cyclin D1. Consequently, apoptosis was triggered by curcumin through the mitochondrial pathway via downregulation of Bcl-2, a downstream anti-apoptotic effector of the Shh signaling. Importantly, the resistant cells that exhibited no decrease in the levels of Shh and Bcl-2, were sensitized to curcumin by the addition of the Shh antagonist, cyclopamine. Furthermore, we have shown that curcumin enhances the killing efficiency of nontoxic doses of cisplatin and gamma-rays. In addition, we present clear evidence that piperine, an enhancer of curcumin bioavailability in humans, potentiates the apoptotic effect of curcumin against medulloblastoma cells. This effect was mediated through strong downregulation of Bcl-2. These results indicate that curcumin, a natural nontoxic compound, represents great promise as Shh-targeted therapy for medulloblastomas.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cerebellar Neoplasms/pathology , Curcumin/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Medulloblastoma/pathology , Signal Transduction/drug effects , Apoptosis/radiation effects , Blotting, Western , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cerebellar Neoplasms/metabolism , Drug Resistance, Neoplasm , Flow Cytometry , Gamma Rays , Humans , Immunoblotting , Immunoenzyme Techniques , Medulloblastoma/metabolism , Mitochondria/drug effects , Mitochondria/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Veratrum Alkaloids/pharmacology
8.
Cancer Res ; 68(8): 2717-25, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413739

ABSTRACT

It has become clear that the initiation and progression of carcinomas depend not only on alterations in epithelial cells, but also on changes in their microenvironment. To identify these changes, we have undertaken cellular and molecular characterization of carcinoma-associated fibroblasts (CAF) and their tumor counterpart fibroblasts (TCF) isolated from 12 breast cancer patients. Normal breast fibroblasts (NBF) from plastic surgery were used as normal control. We present evidence that both CAFs and TCFs are myofibroblasts and show tumor-associated features. Indeed, the p53/p21 response pathway to gamma-rays was defective in 70% CAFs, whereas it was normal in all the TCF and NBF cells. In addition, the basal levels of the p53 and p21 proteins were significantly low in 83% of CAFs and modulated in the majority of TCFs compared with NBFs. Interestingly, both TCFs and CAFs expressed high levels of the cancer marker survivin and consequently exhibited high resistance to cisplatin and UV light. Moreover, most CAFs were positive for the proliferation marker Ki-67 and exhibited high proliferation rate compared with NBFs and TCFs. However, proliferating cell nuclear antigen was highly expressed in both CAFs and TCFs. Using the two-dimensional gel electrophoresis technique, we have also shown that CAF, TCF, and NBF cells present different proteome profiles, with many proteins differentially expressed between these cells. Taken together these results indicate that different genetic alterations can occur in breast CAFs and their corresponding adjacent counterparts, showing the important role that stroma could play in breast carcinogenesis and treatment.


Subject(s)
Breast Neoplasms/pathology , Fibroblasts/pathology , Cell Death , Cell Division , Cell Transformation, Neoplastic , Coloring Agents , Female , Fibroblasts/cytology , Fibroblasts/radiation effects , Flow Cytometry , Gamma Rays , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/analysis , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Reference Values , Survivin , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...