Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Transl Oncol ; 37: 101780, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37672859

ABSTRACT

BACKGROUND: Radiotherapy is an important therapeutic strategy for breast cancer patients through reducing the chances of recurrence and metastasis, which are fueled by cancer-associated fibroblasts (CAFs). Thereby, we addressed here the effect of various doses of X-rays on breast CAFs and their adjacent counterparts. METHODS: We have used WST1 and annexin V-associated with flow cytometry to test the cytotoxic effects of X-rays. Immunoblotting and ELISA was used to assess the expression/secretion of various proteins. Immunohistochemistry was utilized to determine the level of ß-galactosidase and Ki-67. Sphere formation assay was used to test the ability of breast cancer cells to form tumorspheres. Orthotopic tumor xenografts were also used to evaluate the effect of X-ray-treated breast stromal fibroblasts on breast cancer tumor growth in vivo. RESULTS: Breast stromal fibroblasts showed high resistance to X-rays. While the low dose (5 Gy) inhibited cell proliferation and the active features of CAFs, the higher doses (16 and 50 Gy) promoted senescence. However, this was not accompanied by the senescence-associated secretory phenotype (SASP), but rather a reduction in the synthesis/secretion of various cancer-associated cytokines. Additionally, X-rays suppressed the features of active breast stromal fibroblasts, and their paracrine pro-carcinogenic effects. The ablative dose (16 Gy) inhibited the capacity of active stromal fibroblasts to promote the pro-metastatic processes epithelial-to-mesenchymal transition, the formation of cancer stem cells, as well as the growth of humanized orthotopic breast tumor xenografts. CONCLUSION: Together, these findings indicate that X-rays can normalize the features of active breast stromal fibroblasts through promoting senescence without SASP.

2.
Oncogenesis ; 9(8): 70, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32759946

ABSTRACT

The AU-rich element RNA-binding protein 1 (AUF1) is an RNA-binding protein, which can both stabilize and destabilize the transcripts of several cancer-related genes. Since epithelial-to-mesenchymal transition (EMT) and the acquisition of cancer stem cell traits are important for cancer onset and progression, we sought to determine the role of AUF1 in these two important processes. We have shown that AUF1 induces EMT and stemness in breast epithelial cells via stabilization of the SNAIL1 and TWIST1 mRNAs, and their consequent upregulation. Indeed, AUF1 binds the transcripts of these two genes at their 3'UTR and reduces their turnover. Ectopic expression of AUF1 also promoted stemness in mammary epithelial cells, and thereby increased the proportion of cancer stem cells. Importantly, breast cancer cells that ectopically express AUF1 were more efficient in forming orthotopic tumor xenografts in nude mice than their corresponding controls with limiting cell inocula. On the other hand, AUF1 downregulation with specific siRNA inhibited EMT and reduced the stemness features in breast cancer cells. Moreover, AUF1 knockdown sensitized breast cancer cells to the killing effect of cisplatin. Together, these findings provide clear evidence that AUF1 is an important inducer of the EMT process through stabilization of SNAIL1 and TWIST1 and the consequent promotion of breast cancer stem cells. Thereby, AUF1 targeted molecules could constitute efficient therapeutics for breast cancer patients.

3.
Mol Cell Biol ; 40(9)2020 04 13.
Article in English | MEDLINE | ID: mdl-32015100

ABSTRACT

During aging, cellular plasticity and senescence play important roles in tissue regeneration and the pathogenesis of different diseases, including cancer. We have recently shown that senescent breast luminal cells can activate their adjacent stromal fibroblasts. In the present report, we present clear evidence that these senescence-related active fibroblasts can dedifferentiate proliferating primary human luminal cells to multipotent stem cells in an interleukin-8 (IL-8)-dependent manner. This was confirmed using recombinant IL-8, while the truncated protein was not active. This IL-8-related dedifferentiation of luminal cells was mediated through the STAT3-dependent downregulation of p16INK4A and the microRNA miR-141. Importantly, these in vitro-generated mammary stem cells exhibited high molecular and cellular similarities to human mammary stem cells. They have also shown a long-term mammary gland-reconstituting ability and the capacity to produce milk postdelivery. Thereby, these IL-8-generated mammary stem cells could be of great value for autologous cell therapy procedures and also for biomedical research as well as drug development.


Subject(s)
Breast/cytology , Breast/metabolism , Interleukin-8/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Cell Dedifferentiation/physiology , Cell Line, Tumor , Cell Movement/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epithelial-Mesenchymal Transition , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Interleukin-6/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
4.
Oncotarget ; 10(47): 4868-4879, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31448053

ABSTRACT

Osteosarcoma is the most common malignant bone tumor in children, adolescents, and young adults. This pleiomorphic tumor depends on new blood vessel development, also known as angiogenesis, for tumor growth and metastasis. Therefore, it's of utmost importance to identify the key genes and pathways that regulate this pro-metastatic process in order to develop more efficient therapies. Here, we have shown that the RNA-binding protein AUF1 positively regulates the expression of the pro-angiogenic factor VEGF-A and its positive regulator HIF-1alpha through direct binding and stabilization of their mRNAs. This effect is mediated through the seeding sequence of the AUF1 protein in the VEGF-A and HIF-1alpha 3'UTR sequences. As a consequence, the expression of the 3 genes was highly correlative in various osteosarcoma cell lines, and AUF1 enhanced the pro-angiogenic capabilities of osteosarcoma cells both in vitro and in vivo. Indeed, while inhibition of AUF1 using specific siRNA suppressed the pro-angiogenic effects of osteosarcoma cells, ectopic expression of AUF1 enhanced the pro-angiogenic effect in a VEGF-A-dependent manner. Therefore, in the era of targeted therapy, anti-angiogenic therapies targeting AUF1 could provide effective methods for treating osteosarcoma.

5.
Mol Cell Biol ; 39(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30397072

ABSTRACT

Increasing evidence supports the critical role of active stromal adipocytes in breast cancer development and spread. However, the mediators and the mechanisms of action are still elusive. We show here that cancer-associated adipocytes (CAAs) isolated from 10 invasive breast carcinomas are proinflammatory and exhibit active phenotypes, including higher proliferative, invasive, and migratory capacities compared to their adjacent tumor-counterpart adipocytes (TCAs). Furthermore, all CAAs secreted higher level of interleukin-8 (IL-8), which is critical in mediating the paracrine procarcinogenic effects of these cells. Importantly, ectopic expression of IL-8 in TCA cells activated them and enhanced their procarcinogenic effects both in vitro, in a STAT3-dependent manner, and in vivo In contrast, inhibition of the IL-8 signaling using specific short hairpin RNA, anti-IL-8 antibody, or reparixin suppressed the active features of CAAs, including their non-cell-autonomous tumor-promoting activities both on breast luminal cells and in orthotopic tumor xenografts in mice. IL-8 played also an important role in enhancing the proangiogenic effects of breast adipocytes. These results provide clear indication that IL-8 plays key roles in the activation of breast CAAs and acts as a major mediator for their paracrine protumorigenic effects. Thus, targeting CAAs by inhibiting the IL-8 pathway could have great therapeutic value.


Subject(s)
Adipocytes/immunology , Breast Neoplasms/metabolism , Interleukin-8/immunology , Adipocytes/pathology , Angiogenesis Inducing Agents/immunology , Angiogenesis Inducing Agents/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Transformation, Neoplastic , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Heterografts , Humans , MCF-7 Cells , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Primary Cell Culture , STAT3 Transcription Factor/immunology , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology
6.
Mol Cell Biol ; 39(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30397077

ABSTRACT

Aging and stress promote senescence, which has intrinsic tumor suppressor functions and extrinsic tumor promoting properties. Therefore, it is of utmost importance to delineate the effects of senescence inducers on the various types of cells that compose the different organs. We show here that primary normal breast luminal (NBL) cells are more sensitive than their corresponding stromal fibroblasts to proliferative as well as oxidative damage-induced senescence. Like fibroblasts, senescent NBL cells secreted elevated amounts of various cytokines, including interleukin-6 (IL-6) and IL-8, and expressed high levels of p16, p21, and p53, while lamin B1 was downregulated. When senescent, luminal cells activated stromal fibroblasts in an IL-8-dependent manner, through the activation of the STAT3 pathway. These myofibroblasts promoted the epithelial-to-mesenchymal transition and the stemness processes in breast cancer cells in a paracrine manner both in vitro and in a breast cancer animal model. These results show the role of senescent breast luminal cells in promoting the inflammatory/carcinogenic microenvironment through the activation of fibroblasts in an IL-8-dependent manner.


Subject(s)
Breast Neoplasms/metabolism , Interleukin-8/metabolism , Stromal Cells/metabolism , Breast Neoplasms/blood supply , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Transformation, Neoplastic , Cellular Senescence/physiology , Epithelial-Mesenchymal Transition , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Interleukin-6/metabolism , Myofibroblasts/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Primary Cell Culture , STAT3 Transcription Factor/metabolism , Signal Transduction , Stromal Cells/pathology , Tumor Microenvironment , Tumor Suppressor Protein p53/metabolism
7.
Mol Cancer Res ; 16(8): 1299-1308, 2018 08.
Article in English | MEDLINE | ID: mdl-29739873

ABSTRACT

p16INK4A and p53 are two major tumor suppressor proteins that are both upregulated in response to various cellular stresses and during senescence and aging. p53 is a well-characterized transcription factor, while p16INK4A a cyclin-dependent kinase inhibitor encoded by the CDKN2A gene, and controls the expression of several genes through protein-protein interactions and also via miRNAs. This report demonstrates a p16INK4A-dependent positive regulation of p53 expression, at the protein level, in various human cells as well as in mouse embryonic fibroblasts. p16 suppresses p53 turnover through inhibition of its MDM2-related ubiquitination. This effect occurs through p16-related promotion of the MDM2 mRNA turnover via the p16INK4A downstream effectors miR-141 and miR-146b-5p, which bind specific sites at the 3' untranslated region of the MDM2 mRNA.Implications: The current findings show p16INK4A-dependent stabilization of p53 through miR-141/miR-146b-5p-related posttranscriptional repression of MDM2, thus providing new insights into the complex functional link between p16INK4A and p53. Mol Cancer Res; 16(8); 1299-308. ©2018 AACR.


Subject(s)
Genes, p16/physiology , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , Animals , Humans , Mice , Proto-Oncogene Proteins c-mdm2/metabolism , Transfection
8.
Mol Cell Biol ; 37(17)2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28630279

ABSTRACT

Obesity is increasingly recognized as a risk factor for breast cancer development. However, the molecular basis of obesity-related breast carcinogenesis remains elusive. In this study, we have shown that obesity reduces the level of the tumor suppressor p16INK4A protein in breast adipocytes, which showed active features and strong procarcinogenic potential both in vitro and in orthotopic tumor xenografts compared to mature adipocytes from lean women. Furthermore, obesity triggered epithelial-to-mesenchymal transition (EMT) in breast ductal epithelial cells. Interestingly, specific downregulation of p16INK4A increased the expression/secretion levels of various adipokines, including leptin, and activated breast adipocytes from lean women. Consequently, like breast adipocytes from obese women, p16-deficient adipocytes induced EMT in normal primary breast luminal cells in a leptin-dependent manner and enhanced tumor growth. Additionally, we have shown that p16INK4A negatively controls leptin at the mRNA level through microRNAs 141 and 146b-5p (miR-141 and miR-146b-5p), which bind the leptin mRNA at a specific sequence in the 3' untranslated region (UTR). These results show that obesity activates breast stromal adipocytes through p16 downregulation, which upregulates leptin and promotes procarcinogenic processes.


Subject(s)
Adipocytes/metabolism , Breast/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Obesity/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Female , Fibroblasts/metabolism , Humans , Stromal Cells/metabolism
9.
Mol Carcinog ; 56(7): 1687-1702, 2017 07.
Article in English | MEDLINE | ID: mdl-28218424

ABSTRACT

p16INK4A and p53 are two important tumor suppressor proteins that play essential roles during cell proliferation and aging through regulating the expression of several genes. Here, we report that p16INK4A and p53 co-regulate a plethora of transcripts. Furthermore, both proteins colocalize in the nucleus of human primary skin fibroblasts and breast luminal cells, and form a heteromer whose level increases in response to genotoxic stress as well as aging of human fibroblasts and various mouse organs. CDK4 is also present in this heteromeric complex, which is formed only in the presence of DNA both in vitro using pure recombinant proteins and in vivo. We have also shown that p16INK4A enhances the binding efficiency of p53 to its cognate sequence presents in the CDKN1A promoter in vitro, and both proteins are present at the promoters of CDKN1A and BAX in vivo. Importantly, the fourth ankyrin repeat of p16INK4A and the C-terminal domain of p53 were necessary for the physical association between these two proteins. The physiologic importance of this association was revealed by the inability of cancer-associated p16INK4A mutants to interact with p53 and to transactivate the expression of its major targets CDKN1A and BAX in the p16-defective U2OS cells expressing either wild-type or mutated p16INK4A . Furthermore, the association between p16INK4A and p53 was capital for their nuclear colocalization, the X-ray-dependent induction of p21 and Bax proteins as well as the induction of apoptosis in various types of cells. Together, these results show DNA-dependent physical interaction between p16INK4A and p53.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA/metabolism , Gene Expression Regulation , Tumor Suppressor Protein p53/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/metabolism
10.
Mol Carcinog ; 56(3): 985-999, 2017 03.
Article in English | MEDLINE | ID: mdl-27596953

ABSTRACT

Senescence and epithelial-to-mesenchymal transition (EMT) processes are under the control of common tumor suppressor proteins, EMT transcription factors, and microRNAs. However, the molecular mechanisms that coordinate the functional link between senescence and EMT are still elusive. We have shown here that p16INK4A -related induction of senescence is mediated through miR-141 and miR-146b-5p. These two microRNAs are up-regulated in aging human fibroblast and epithelial cells. Furthermore, miR-141 and miR146b-5p trigger cell cycle arrest at G1 phase and induce senescence in primary human fibroblasts and breast cancer cells in the presence and absence of p16INK4A . Like p16INK4A -induced senescence, miR-141/miR146b-5p-related senescence is not associated with secretory phenotype, and is mediated through the RNA binding protein AUF1. We have further demonstrated that p16INK4A and its downstream miRNA targets inhibit EMT through suppressing the EMT inducer ZEB1 in an AUF1-dependent manner. Indeed, AUF1 binds the mRNA of this gene leading to increase in its level. These results indicate that p16INK4A controls both senescence and EMT through repressing EMT-related transcription factor via miR-141/miR146b-5p and their target AUF1. This sheds more light on the molecular basis of the tumor suppressive functions of p16INK4A , which represses both the proliferative and the migratory/invasive capacities of cells. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p18/genetics , Epithelial Cells/cytology , Fibroblasts/cytology , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , MicroRNAs/genetics , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Up-Regulation
11.
BMC Cancer ; 16: 540, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27465411

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive histological subtype with limited treatment options and very poor prognosis following progression after standard chemotherapeutic regimens. Therefore, novel molecules and therapeutic options are urgently needed for this category of patients. Recently, we have identified PAC as a curcumin analogue with potent anti-cancer features. METHODS: HPLC was used to evaluate the stability of PAC and curcumin in PBS and also in circulating blood. Cytotoxicity/apoptosis was assessed in different breast cancer cell lines using propidium iodide/annexinV associated with flow cytometry. Furthermore, immunoblotting analysis determined the effects of PAC on different oncogenic proteins and pathways. Additionally, the real time xCELLigence RTCA technology was applied to investigate the effect of PAC on the cellular proliferation, migration and invasion capacities. RESULTS: PAC is more stable than curcumin in PBS and in circulating blood. Furthermore, we have shown differential sensitivity of estrogen receptor-alfa positive (ERα(+)) and estrogen receptor alfa negative (ERα(-)) breast cancer cells to PAC, which down-regulated ERα in both cell types. This led to complete disappearance of ERα in ERα(-) cells, which express very low level of this receptor. Interestingly, specific down-regulation of ERα in receptor positive cells increased the apoptotic response of these cells to PAC, confirming that ERα inhibits PAC-dependent induction of apoptosis, which could be mediated through ERα down-regulation. Additionally, PAC inhibited the proliferation and suppressed the epithelial-to-mesenchymal transition process in breast cancer cells, with higher efficiency on the TNBC subtype. This effect was also observed in vivo on tumor xenografts. Additionally, PAC suppressed the expression/secretion of 2 important cytokines IL-6 and MCP-1, and consequently inhibited the paracrine procarcinogenic effects of breast cancer cells on breast stromal fibroblasts. CONCLUSION: These results indicate that PAC could be considered as important candidate for future therapeutic options against the devastating TNBC subtype.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzylidene Compounds/administration & dosage , Epithelial-Mesenchymal Transition/drug effects , Estrogen Receptor alpha/metabolism , Piperidones/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemokine CCL2/metabolism , Curcumin/administration & dosage , Curcumin/analogs & derivatives , Curcumin/pharmacology , Drug Stability , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/metabolism , Mice , Piperidones/pharmacology , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
12.
Mol Carcinog ; 55(3): 233-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25641341

ABSTRACT

Colorectal cancer (CRC) is a major cause of cancer morbidity and mortality worldwide. Although response rates and overall survival have been improved in recent years, resistance to multiple drug combinations is inevitable. Therefore, the development of more efficient drugs, with fewer side effects is urgently needed. To this end, we have investigated in the present report the effect of PAC, a novel cucumin analogue, on CRC cells both in vitro and in vivo. We have shown that PAC induces apoptosis, mainly via the internal mitochondrial route, and inhibits cell proliferation through delaying the cell cycle at G2/M phase. Interestingly, the pro-apoptotic effect was mediated through STAT3-dependent down-regulation of cyclin D1 and its downstream target survivin. Indeed, change in the expression level of cyclin D1 modulated the expression of survivin and the response of CRC cells to PAC. Furthermore, using the ChIP assay, we have shown PAC-dependent reduction in the binding of STAT3 to the cyclin D1 promoter in vivo. Additionally, PAC suppressed the epithelial-to-mesenchymal process through down-regulating the mesenchymal markers (N-cadherin, vimentin and Twist1) and inhibiting the invasion/migration abilities of the CRC cells via repressing the pro-migration/invasion protein kinases AKT and ERK1/2. In addition, PAC inhibited tumor growth and repressed the JAK2/STAT3, AKT/mTOR and MEK/ERK pathways as well as their common downstream effectors cyclin D1 and survivin in humanized CRC xenografts. Collectively, these results indicate that PAC has potent anti-CRC effects, and therefore could constitute an effective alternative chemotherapeutic agent, which may consolidate the adjuvant treatment of colon cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Colon/drug effects , Colorectal Neoplasms/drug therapy , Curcumin/therapeutic use , Cyclin D1/metabolism , Epithelial-Mesenchymal Transition/drug effects , Rectum/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Curcumin/analogs & derivatives , Curcumin/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , Mice, Nude , Rectum/metabolism , Rectum/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
13.
J Biol Chem ; 289(45): 31433-47, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25261470

ABSTRACT

miR-141 and miR-146b-5p are two important tumor suppressor microRNAs, which control several cancer-related genes and processes. In the present report, we have shown that these microRNAs bind specific sites at the 3'-untranslated region (UTR) of the mRNA-binding protein AUF1, leading to its down-regulation. This inverse correlation between the levels of these microRNAs and AUF1 has been identified in various osteosarcoma cell lines. Additionally, we present clear evidence that AUF1 promotes mesenchymal features in osteosarcoma cells and that miR-141 and miR-146b-5p suppress this prometastatic process through AUF1 repression. Indeed, both microRNAs suppressed the invasion/migration and proliferation abilities of osteosarcoma cells through inhibiting the AKT protein kinase in an AUF1-dependent manner. We have also shown that AUF1 binds to and stabilizes the mRNA of the AKT activator phosphoinositide-dependent kinase-1 (PDK1). Furthermore, miR-141 and miR-146b-5p positively regulate the epithelial markers (E-cadherin and Epcam) and repress the mesenchymal markers (N-cadherin, Vimentin, Twist2, and ZEB1). These effects were mediated via the repression of the epithelial-to-mesenchymal inducer ZEB1 through targeting AUF1, which binds the 3'-UTR of the ZEB1 mRNA and reduces its turnover. These results indicate that at least some tumor suppressor functions of miR-141 and miR-146b-5p are mediated through the repression of the oncogenic potentials of AUF1. Therefore, these 3'-UTR-directed post-transcriptional gene expression regulators constitute promising new targets for diagnostic and/or therapeutic interventions.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Homeodomain Proteins/metabolism , MicroRNAs/physiology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Base Sequence , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Humans , Mesoderm/cytology , Molecular Sequence Data , Neoplasm Metastasis , Neoplasms/metabolism , Prognosis , Protein Binding , Sequence Homology, Nucleic Acid , Zinc Finger E-box-Binding Homeobox 1
14.
J Biol Chem ; 289(45): 30962-76, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25231991

ABSTRACT

The development and spread of mammary carcinomas require synergetic interplay between tumor cells and their microenvironment through paracrine secretions, which are still not well defined. We have shown here that interleukin-6 (IL-6), either recombinant or secreted from highly invasive breast cancer cells, down-regulates the tumor suppressor proteins p16(INK4A), p21(WAF1), and p53 and activates breast stromal fibroblasts in a paracrine manner. The formation of myofibroblasts requires p16(INK4A) down-regulation and the activation of the JAK2/STAT3 pathway. Indeed, the transcription factor STAT3 positively controls the expression of the three major myofibroblast markers, SDF-1, α-smooth muscle actin (α-SMA), and TGF-ß1, and mediates IL-6-related down-regulation of p16(INK4A), p21(WAF1), and p53 as well as the activation of stromal fibroblasts. Importantly, these effects were mediated through STAT3-dependent up-regulation of the mRNA-binding protein AUF1, whose promoter contains three canonical STAT3 binding sites. AUF1 binds the SDF-1, α-SMA, TGF-ß1, and IL-6 mRNAs and reduces their turnover. Consequently, specific AUF1 down-regulation inhibits IL-6-dependent activation of breast stromal fibroblasts, whereas AUF1 ectopic expression of p37(AUF1) activated these cells and enhanced their paracrine induction of epithelial-to-mesenchymal transition in breast cancer cells, which shows a non-cell-autonomous oncogenic function of AUF1. Together, these results demonstrate a major role of IL-6 in activating breast stromal fibroblasts through STAT3-dependent AUF1 induction.


Subject(s)
Fibroblasts/cytology , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Stromal Cells/cytology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Culture Media, Conditioned/chemistry , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Fibroblasts/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Myofibroblasts/metabolism , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
15.
Mol Carcinog ; 53(12): 979-87, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23813879

ABSTRACT

Ultraviolet (UV) light is a carcinogenic agent that upregulates the expression of several genes involved in various cellular processes, including cell cycle checkpoints and apoptosis. The universal cyclin-dependent kinase inhibitor p21(WAF1/Cip1) plays major roles in these processes, and the level of its corresponding message increases several times in response to UV-induced DNA damage. This upregulation is mainly posttranscriptional owing to HuR-dependent mRNA stabilization. Since the protein kinase Atr plays major roles during the cellular response to UV damage, we sought to investigate its possible implication in the stabilization of the p21(WAF1/Cip1) coding mRNA. We have shown that the UV-dependent accumulation of the CDKN1A mRNA is indeed under the control of the Atr protein kinase. Upon UV damage, Atr allows nuclear-cytoplasmic shuttling of the HuR protein, which binds the CDKN1A mRNA and reduces its turnover. This ATR-dependent effect is mediated through UV-related phosphorylation/inactivation of the Cdk1 protein kinase by Atr, which leads to the dissociation of HuR from Cdk1. Indeed, inhibition or shRNA specific knockdown of CDK1 in ATR-deficient cells enhanced the cytoplasmic level of HuR and restored the CDKN1A mRNA upregulation in response to UV damage. These results show that ATR stabilizes the CDKN1A message in response to UV damage through Cdk1-related cytoplasmic accumulation of HuR.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinases/genetics , ELAV Proteins/genetics , RNA, Messenger/genetics , Up-Regulation/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , CDC2 Protein Kinase , Cell Cycle Proteins/genetics , Cell Line , Cytoplasm/genetics , DNA Damage/genetics , ELAV-Like Protein 1 , Humans , Phosphorylation/genetics , Transcriptional Activation/genetics , Ultraviolet Rays
16.
J Biol Chem ; 288(49): 35511-25, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24163379

ABSTRACT

p16(INK4a) is a tumor suppressor protein involved in several stress-related cellular responses, including apoptosis. Recent lines of evidence indicate that p16(INK4a) is also a modulator of gene expression. However, the molecular mechanisms underlying this novel function are still obscure. Here, we present clear evidence that p16(INK4a) modulates the levels of various microRNAs, with marked positive effect on miR-141 and miR-146b-5p. This effect is mediated through the formation of the p16-CDK4-Sp1 heterocomplex, which binds to Sp1 consensus-binding motifs present in the promoters of miR-141 and miR-146b-5p, and it enables their transcription. In addition, we have shown that p16(INK4a) interacts with Sp1 through the fourth ankyrin repeat, which is crucial for Sp1 binding to the miR-141 and miR-146b-5p promoters and their transcriptional activation. The physiological importance of this association was revealed by the inability of cancer-related p16(INK4a) mutants to interact with Sp1. Moreover, we have shown p16-CDK4-Sp1-dependent up-regulation of miR-141 and miR-146b-5p following UV light-induced DNA damage and the role of these two microRNAs in mediating p16-related induction of apoptosis in response to this genotoxic stress. Together, these results indicate that p16(INK4a) associates with CDK4 not only to inhibit the cell cycle but also to enable the transcription of two important onco-microRNAs, which act as downstream effectors.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Sp1 Transcription Factor/metabolism , Base Sequence , Cell Cycle , Cell Line , Cyclin-Dependent Kinase 4/chemistry , Cyclin-Dependent Kinase Inhibitor p16/chemistry , DNA Damage , Humans , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Promoter Regions, Genetic , Protein Binding , RNA Stability , Sp1 Transcription Factor/chemistry , Transcriptional Activation/radiation effects , Ultraviolet Rays/adverse effects
17.
PLoS One ; 8(7): e70133, 2013.
Article in English | MEDLINE | ID: mdl-23894605

ABSTRACT

BACKGROUND: p16(INK4a) and p21(WAF1) are two independent cyclin-dependent kinase inhibitors encoded by the CDKN2A and CDKN1A genes, respectively. p16(INK4a) and p21(WAF1) are similarly involved in various anti-cancer processes, including the regulation of the critical G1 to S phase transition of the cell cycle, senescence and apoptosis. Therefore, we sought to elucidate the molecular mechanisms underlying the link between these two important tumor suppressor proteins. METHODOLOGY/PRINCIPAL FINDINGS: We have shown here that the p16(INK4a) protein positively controls the expression of p21(WAF1) in both human and mouse cells. p16(INK4a) stabilizes the CDKN1A mRNA through negative regulation of the mRNA decay-promoting AUF1 protein. Immunoprecipitation of AUF1-associated RNAs followed by quantitative RT-PCR indicated that endogenous AUF1 binds to the CDKN1A mRNA in a p16(INK4A)-dependent manner. Furthermore, while AUF1 down-regulation increased the expression level of the CDKN1A mRNA, the concurrent knockdown of AUF1 and CDKN2A, using specific silencing RNAs, restored the normal expression of the gene. Moreover, we used EGFP reporter fused to the CDKN2A AU-rich element (ARE) to demonstrate that p16(INK4A) regulation of the CDKN1A mRNA is AUF1- and ARE-dependent. Furthermore, ectopic expression of p16(INK4A) in p16(INK4A)-deficient breast epithelial MCF-10A cells significantly increased the level of p21(WAF1), with no effect on cell proliferation. In addition, we have shown direct correlation between p16(INK4a) and p21(WAF1) levels in various cancer cell lines. CONCLUSION/SIGNIFICANCE: These findings show that p16(INK4a) stabilizes the CDKN1A mRNA in an AUF1-dependent manner, and further confirm the presence of a direct link between the 2 important cancer-related pathways, pRB/p16(INK4A) and p14(ARF)/p53/p21(WAF1).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation , Animals , Cell Line, Tumor , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Mice , RNA Processing, Post-Transcriptional , RNA Stability , Signal Transduction
18.
Neoplasia ; 15(6): 631-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23730211

ABSTRACT

Activated cancer-associated fibroblasts (CAFs) or myofibroblasts not only facilitate tumor growth and spread but also affect tumor response to therapeutic agents. Therefore, it became clear that efficient therapeutic regimens should also take into account the presence of these supportive cells and inhibit their paracrine effects. To this end, we tested the effect of low concentrations of curcumin, a pharmacologically safe natural product, on patient-derived primary breast CAF cells. We have shown that curcumin treatment upregulates p16(INK4A) and other tumor suppressor proteins while inactivates the JAK2/STAT3 pathway. This reduced the level of alpha-smooth muscle actin (α-SMA) and the migration/invasion abilities of these cells. Furthermore, curcumin suppressed the expression/secretion of stromal cell-derived factor-1 (SDF-1), interleukin-6 (IL-6), matrix metalloproteinase-2 (MMP-2), MMP-9, and transforming growth factor-ß, which impeded their paracrine procarcinogenic potential. Intriguingly, these effects were sustained even after curcumin withdrawal and cell splitting. Therefore, using different markers of senescence [senescence-associated ß-galactosidase (SA-ß-gal) activity, Ki-67 and Lamin B1 levels, and bromodeoxyuridine incorporation], we have shown that curcumin markedly suppresses Lamin B1 and triggers DNA damage-independent senescence in proliferating but not quiescent breast stromal fibroblasts. Importantly, this curcumin-related senescence was p16(INK4A)-dependent and occurred with no associated inflammatory secretory phenotype. These results indicate the possible inactivation of cancer-associated myofibroblasts and present the first indication that curcumin can trigger DNA damage-independent and safe senescence in stromal fibroblasts.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Curcumin/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Fibroblasts/drug effects , Actins/metabolism , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cellular Senescence/drug effects , DNA Damage/drug effects , Dose-Response Relationship, Drug , Female , Fibroblasts/cytology , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Humans , Interleukin-6/metabolism , Ki-67 Antigen/metabolism , Lamin Type B/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , STAT3 Transcription Factor/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Up-Regulation
19.
Age (Dordr) ; 35(3): 549-62, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22252435

ABSTRACT

Survivin, an important anti-apoptotic protein, is highly expressed in most cancers, which generally arise in cells of older individuals. We have shown here accumulation of survivin and phospho-survivin in aged normal human skin fibroblasts and mice organs. This age-related accumulation of survivin was due to protein stabilization through association with the molecular chaperone Hsp90 protein, which was also up-regulated during aging. Interestingly, Hsp90 binds preferentially to phospho-survivin, which explains its higher stability. In addition, we provide clear evidence that aged cells exhibit apoptosis resistance when challenged with UV light, cisplatin, γ-rays or H2O2 as compared to their younger counterparts. In response to γ-rays and H2O2, the levels of Bcl-2 and both forms of survivin were up-regulated in old cells, but not in their corresponding young ones. This repression of survivin and phospho-survivin in young cells is p53 dependent. Importantly, survivin inhibition/down-regulation with flavopiridol or specific shRNAs increased the apoptotic response of old fibroblasts to various genotoxic agents, and restored the pro-apoptotic Bax/Bcl2 ratio and the increase in the levels of cleaved caspase-3 and PARP in old cells. These results show the role of survivin in the age-dependent resistance of human fibroblasts, and provide new insights into the molecular mechanisms that underlie the complex relationship between aging, apoptosis, and cancer.


Subject(s)
Aging/genetics , Apoptosis/genetics , DNA Damage/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Inhibitor of Apoptosis Proteins/genetics , RNA/genetics , Repressor Proteins/genetics , Aging/metabolism , Aging/pathology , Animals , Disease Models, Animal , Female , Fibroblasts/pathology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Inhibitor of Apoptosis Proteins/biosynthesis , Mice , Mice, Inbred C57BL , RNA/metabolism , Repressor Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Survivin
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...