Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 36(23): 3322-3333, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28068326

ABSTRACT

Melanoma is the most lethal form of skin cancer and treatment of metastatic melanoma remains challenging. BRAF/MEK inhibitors show only temporary benefit due the occurrence of resistance and immunotherapy is effective only in a subset of patients. To improve patient survival, there is a need to better understand molecular mechanisms that drive melanoma growth and operate downstream of the mitogen activated protein kinase (MAPK) signaling. The Krüppel-like factor 4 (KLF4) is a zinc-finger transcription factor that plays a critical role in embryonic development, stemness and cancer, where it can act either as oncogene or tumor suppressor. KLF4 is highly expressed in post-mitotic epidermal cells, but its role in melanoma remains unknown. Here, we address the function of KLF4 in melanoma and its interaction with the MAPK signaling pathway. We find that KLF4 is highly expressed in a subset of human melanomas. Ectopic expression of KLF4 enhances melanoma cell growth by decreasing apoptosis. Conversely, knock-down of KLF4 reduces melanoma cell proliferation and induces cell death. In addition, depletion of KLF4 reduces melanoma xenograft growth in vivo. We find that the RAS/RAF/MEK/ERK signaling positively modulates KLF4 expression through the transcription factor E2F1, which directly binds to KLF4 promoter. Overall, our data demonstrate the pro-tumorigenic role of KLF4 in melanoma and uncover a novel ERK1/2-E2F1-KLF4 axis. These findings identify KLF4 as a possible new molecular target for designing novel therapeutic treatments to control melanoma growth.


Subject(s)
E2F1 Transcription Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Kruppel-Like Transcription Factors/metabolism , Melanoma/pathology , raf Kinases/metabolism , ras Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , E2F1 Transcription Factor/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/genetics , ras Proteins/genetics
2.
Cell Death Differ ; 22(12): 2006-19, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26024388

ABSTRACT

HEDGEHOG (HH) signaling is a key regulator of tissue development and its aberrant activation is involved in several cancer types, including melanoma. We and others have shown a reciprocal cross talk between HH signaling and p53, whose function is often impaired in melanoma. Here we present evidence that both GLI1 and GLI2, the final effectors of HH signaling, regulate the transcription factor E2F1 in melanoma cells, by binding to a functional non-canonical GLI consensus sequence. Consistently, we find a significant correlation between E2F1 and PATCHED1 (PTCH1), GLI1 and GLI2 expression in human melanomas. Functionally, we find that E2F1 is a crucial mediator of HH signaling and it is required for melanoma cell proliferation and xenograft growth induced by activation of the HH pathway. Interestingly, we present evidence that the HH/GLI-E2F1 axis positively modulates the inhibitor of apoptosis-stimulating protein of p53 (iASPP) at multiple levels. HH activation induces iASPP expression through E2F1, which directly binds to iASPP promoter. HH pathway also contributes to iASPP function, by the induction of Cyclin B1 and by the E2F1-dependent regulation of CDK1, which are both involved in iASPP activation. Our data show that activation of HH signaling enhances proliferation in presence of E2F1 and promotes apoptosis in its absence or upon CDK1 inhibition, suggesting that E2F1/iASPP dictates the outcome of HH signaling in melanoma. Together, these findings identify a novel HH/GLI-E2F1-iASPP axis that regulates melanoma cell growth and survival, providing an additional mechanism through which HH signaling restrains p53 proapoptotic function.


Subject(s)
E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic/genetics , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Base Sequence , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin B1/metabolism , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Mutagenesis, Site-Directed , NIH 3T3 Cells , Oncogene Proteins/genetics , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Repressor Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
3.
Oncogene ; 33(38): 4697-708, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-24681955

ABSTRACT

Melanoma is one of the most aggressive types of human cancer, characterized by enhanced heterogeneity and resistance to conventional therapy at advanced stages. We and others have previously shown that HEDGEHOG-GLI (HH-GLI) signaling is required for melanoma growth and for survival and expansion of melanoma-initiating cells (MICs). Recent reports indicate that HH-GLI signaling regulates a set of genes typically expressed in embryonic stem cells, including SOX2 (sex-determining region Y (SRY)-Box2). Here we address the function of SOX2 in human melanomas and MICs and its interaction with HH-GLI signaling. We find that SOX2 is highly expressed in melanoma stem cells. Knockdown of SOX2 sharply decreases self-renewal in melanoma spheres and in putative melanoma stem cells with high aldehyde dehydrogenase activity (ALDH(high)). Conversely, ectopic expression of SOX2 in melanoma cells enhances their self-renewal in vitro. SOX2 silencing also inhibits cell growth and induces apoptosis in melanoma cells. In addition, depletion of SOX2 progressively abrogates tumor growth and leads to a significant decrease in tumor-initiating capability of ALDH(high) MICs upon xenotransplantation, suggesting that SOX2 is required for tumor initiation and for continuous tumor growth. We show that SOX2 is regulated by HH signaling and that the transcription factors GLI1 and GLI2, the downstream effectors of HH-GLI signaling, bind to the proximal promoter region of SOX2 in primary melanoma cells. In functional studies, we find that SOX2 function is required for HH-induced melanoma cell growth and MIC self-renewal in vitro. Thus SOX2 is a critical factor for self-renewal and tumorigenicity of MICs and an important mediator of HH-GLI signaling in melanoma. These findings could provide the basis for novel therapeutic strategies based on the inhibition of SOX2 for the treatment of a subset of human melanomas.


Subject(s)
Melanoma/metabolism , Neoplastic Stem Cells/physiology , SOXB1 Transcription Factors/physiology , Skin Neoplasms/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Apoptosis , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Signal Transduction , Skin Neoplasms/pathology , Transcription Factors/metabolism , Zinc Finger Protein GLI1
4.
Oncogene ; 32(40): 4737-47, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23146903

ABSTRACT

The Hedgehog-GLI (HH-GLI) signaling plays a critical role in controlling growth and tissue patterning during embryogenesis and is implicated in a variety of human malignancies, including those of the skin. Phosphorylation events have been shown to regulate the activity of the GLI transcription factors, the final effectors of the HH-GLI signaling pathway. Here, we show that WIP1 (or PPM1D), an oncogenic phosphatase amplified/overexpressed in several types of human cancer, is a positive modulator of the HH signaling. Mechanistically, WIP1 enhances the function of GLI1 by increasing its transcriptional activity, nuclear localization and protein stability, but not of GLI2 nor GLI3. We also find that WIP1 and GLI1 are in a complex. Modulation of the transcriptional activity of GLI1 by WIP1 depends on the latter's phosphatase activity and, remarkably, does not require p53, a known WIP1 target. Functionally, we find that WIP1 is required for melanoma and breast cancer cell proliferation and self-renewal in vitro and melanoma xenograft growth induced by activation of the HH signaling. Pharmacological blockade of the HH pathway with the SMOOTHENED antagonist cyclopamine acts synergistically with inhibition of WIP1 in reducing growth of melanoma and breast cancer cells in vitro. Overall, our data uncover a role for WIP1 in modulating the activity of GLI1 and in sustaining cancer cell growth and cancer stem cell self-renewal induced by activation of the HH pathway. These findings open a novel therapeutic approach for human melanomas and, possibly, other cancer types expressing WIP1 and with activated HH pathway.


Subject(s)
Hedgehog Proteins/metabolism , Phosphoprotein Phosphatases/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Cell Nucleus/metabolism , Humans , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Phosphatase 2C , Transcription Factors/metabolism , Transcription, Genetic , Zinc Finger Protein GLI1
SELECTION OF CITATIONS
SEARCH DETAIL
...