Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Oncogene ; 35(12): 1602-8, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26119942

ABSTRACT

Metastasis is a multistep cell-biological process, which is orchestrated by many factors, including metastasis activators and suppressors. Metastasis Suppressor 1 (MTSS1) was originally identified as a metastasis suppressor protein whose expression is lost in metastatic bladder and prostate carcinomas. However, recent findings indicate that MTSS1 acts as oncogene and pro-migratory factor in melanoma tumors. Here, we identify and characterized a molecular mechanism controlling MTSS1 expression, which impinges on a pro-tumorigenic role of MTSS1 in breast tumors. We found that in normal and in cancer cell lines ΔNp63 is able to drive the expression of MTSS1 by binding to a p63-binding responsive element localized in the MTSS1 locus. We reported that ΔNp63 is able to drive the migration of breast tumor cells by inducing the expression of MTSS1. Notably, in three human breast tumors data sets the MTSS1/p63 co-expression is a negative prognostic factor on patient survival, suggesting that the MTSS1/p63 axis might be functionally important to regulate breast tumor progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/physiology , Microfilament Proteins/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Transcription, Genetic , Humans
3.
Cell Death Dis ; 6: e2010, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633716

ABSTRACT

Neuroblastoma (NB) is the most common extracranial childhood tumor classified in five stages (1, 2, 3, 4 and 4S), two of which (3 and 4) identify chemotherapy-resistant, highly aggressive disease. High-risk NB frequently displays MYCN amplification, mutations in ALK and ATRX, and genomic rearrangements in TERT genes. These NB subtypes are also characterized by reduced susceptibility to programmed cell death induced by chemotherapeutic drugs. The latter feature is a major cause of failure in the treatment of advanced NB patients. Thus, proper reactivation of apoptosis or of other types of programmed cell death pathways in response to treatment is relevant for the clinical management of aggressive forms of NB. In this short review, we will discuss the most relevant genomic rearrangements that define high-risk NB and the role that destabilization of p53 and p73 can have in NB aggressiveness. In addition, we will propose a strategy to stabilize p53 and p73 by using specific inhibitors of their ubiquitin-dependent degradation. Finally, we will introduce necroptosis as an alternative strategy to kill NB cells and increase tumor immunogenicity.


Subject(s)
Apoptosis/genetics , Neuroblastoma/genetics , Neuroblastoma/therapy , Animals , Cell Proliferation/genetics , Humans , Necrosis/genetics , Neuroblastoma/pathology , Signal Transduction
4.
Oncogene ; 32(6): 797-802, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-22469988

ABSTRACT

Tumor cells activate pathways that facilitate and stimulate glycolysis even in the presence of adequate levels of oxygen in order to satisfy their continuous need of molecules, such as nucleotides, ATP and fatty acids, necessary to support their rapid proliferation. Accordingly, a variety of human tumors are characterized by elevated expression levels of the hexokinase 2 isoform (HK2). Although different molecular mechanisms, including genetic and epigenetic mechanisms, have been suggested to account for the altered expression of HK2 in tumors, the potential role of microRNAs (miRNAs) in the regulation of HK2 expression has not been evaluated. Here, we report that miR-143 inhibits HK2 expression via a conserved miR-143 recognition motif located in the 3'-untranslated region (3'UTR) of HK2 mRNA. We demonstrate that miR143 inhibits HK2 expression both in primary keratinocytes and in head and neck squamous cell carcinoma (HNSCC)-derived cell lines. Importantly, we found that miR-143 inversely correlates with HK2 expression in HNSCC-derived cell lines and in primary tumors. We also report that the miRNA-dependent regulation of hexokinase expression is not limited to HK2 as miR-138 targets HK1 via a specific recognition motif located in its 3'UTR. All these data unveil a new miRNA-dependent mechanism of regulation of hexokinase expression potentially important in the regulation of glucose metabolism of cancer cells.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Hexokinase/genetics , MicroRNAs/physiology , Cell Line, Tumor , Humans , Keratinocytes/metabolism
5.
Oncogene ; 32(39): 4721-6, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23085759

ABSTRACT

The Cullin4A (cul4A)-dependent ligase (CDL4A) E3 has been implicated in a variety of biological processes, including cell cycle progression and DNA damage response. Remarkably, CDL4A exerts its function through both proteolytic and non-proteolytic events. Here, we show that the p53 family member p73 is able to interact with the CDL4A complex through its direct binding to the receptor subunit DNA-binding protein 1 (DDB1). As a result, the CDL4A complex is able to monoubiquitylate p73. Modification of p73 by CDL4A-mediated ubiquitylation does not affect p73 protein stability, but negatively regulates p73-dependent transcriptional activity. Indeed, genetic or RNA interference-mediated depletion of DDB1 induces the expression of several p73 target genes in a p53-independent manner. In addition, by exploiting a bioinformatic approach, we found that elevated expression of Cul4A in human breast carcinomas is associated with repression of p73 target genes. In conclusion, our findings add a novel insight into the regulation of p73 by the CDL4A complex, through the inhibition of its transcriptional function.


Subject(s)
Cullin Proteins/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Nuclear Proteins/antagonists & inhibitors , Transcription, Genetic , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Carrier Proteins/physiology , Cell Line , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , HEK293 Cells , Humans , Mice , Multiprotein Complexes , Neoplasm Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Stability , Transcription, Genetic/physiology , Tumor Protein p73 , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitination
6.
Oncogene ; 29(9): 1384-93, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-19966869

ABSTRACT

Ubiquitin-specific proteases (USPs) are a subclass of cysteine proteases that catalyze the removal of ubiquitin (either monomeric or chains) from substrates, thus counteracting the activity of E3 ubiquitin ligases. Although the importance of USPs in a multitude of processes, from hereditary cancer to neurodegeneration, is well established, our knowledge on their mode of regulation, substrate specificity and biological function is quite limited. In this study we identify USP47 as a novel interactor of the E3 ubiquitin ligase, Skp1/Cul1/F-box protein beta-transducin repeat-containing protein (SCF(beta-Trcp)). We found that both beta-Trcp1 and beta-Trcp2 bind specifically to USP47, and point mutations in the beta-Trcp WD-repeat region completely abolished USP47 binding, indicating an E3-substrate-type interaction. However, unlike canonical beta-Trcp substrates, USP47 protein levels were neither affected by silencing of beta-Trcp nor modulated in a variety of processes, such as cell-cycle progression, DNA damage checkpoint responses or tumor necrosis factor (TNF) pathway activation. Notably, genetic or siRNA-mediated depletion of USP47 induced accumulation of Cdc25A, decreased cell survival and augmented the cytotoxic effects of anticancer drugs. In conclusion, we showed that USP47, a novel beta-Trcp interactor, regulates cell growth and survival, potentially providing a novel target for anticancer therapies.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Survival/physiology , Endopeptidases/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Cell Line, Tumor , Cysteine Proteases/metabolism , F-Box-WD Repeat-Containing Protein 7 , Humans , NF-kappa B/metabolism , RNA, Small Interfering/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/physiology , Transducin , Ubiquitin-Specific Proteases
7.
Oncogene ; 28(35): 3157-66, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19581926

ABSTRACT

The transcription factor p73, a member of the p53 family, mediates cell-cycle arrest and apoptosis in response to DNA damage-induced cellular stress, acting thus as a proapoptotic gene. Similar to p53, p73 activity is regulated by post-translational modification, including phosphorylation, acetylation and ubiquitylation. In C. elegans, the F-box protein FSN-1 controls germline apoptosis by regulating CEP-1, the single ancestral p53 family member. Here we report that FBXO45, the human ortholog of FSN-1, binds specifically to p73 triggering its proteasome-dependent degradation. Importantly, SCF(FBXO45) ubiquitylates p73 both in vivo and in vitro. Moreover, siRNA-mediated depletion of FBXO45 stabilizes p73 and concomitantly induces cell death in a p53-independent manner. All together, these results show that the orphan F-box protein FBXO45 regulates the stability of p73, highlighting a conserved pathway evolved from nematode to human by which the p53 members are regulated by an SCF-dependent mechanism.


Subject(s)
DNA-Binding Proteins/metabolism , F-Box Proteins/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/pathology , CHO Cells , Cell Death/genetics , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cricetinae , Cricetulus , F-Box Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hemagglutinins/metabolism , Humans , Kidney/cytology , Leupeptins/pharmacology , Mutation , Neuroblastoma/pathology , Proteasome Inhibitors , Protein Binding , RNA, Small Interfering/metabolism , Substrate Specificity , Temperature , Transfection , Tumor Protein p73 , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Cell Death Differ ; 15(7): 1103-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552861

ABSTRACT

The HECT-type E3 ubiquitin ligase (E3) Itch is absent in the non-agouti-lethal 18H or Itchy mice, which develop a severe immunological disease, including lung and stomach inflammation and hyperplasia of lymphoid and hematopoietic cells. The involvement of Itch in multiple signaling pathways and pathological conditions is presently an area of extensive scientific interest. This review aims to bring together a growing body of work exploring Itch-regulated biological processes, and to highlight recent discoveries on the regulatory mechanisms modulating its catalytic activity and substrate recognition capability. Our contribution is also an endeavor to correlate Itch substrate specificity with the pathological defects manifested by the mutant Itchy mice.


Subject(s)
Immune System/metabolism , Neoplasms/enzymology , Repressor Proteins/metabolism , Skin/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Death , ErbB Receptors/metabolism , Immune System/pathology , Keratinocytes/metabolism , Mice , Mice, Mutant Strains , Neoplasms/immunology , Neoplasms/pathology , Phosphorylation , Protein Transport , Receptors, Chemokine/metabolism , Repressor Proteins/immunology , Signal Transduction , Skin/immunology , Skin/pathology , Substrate Specificity , TRPC Cation Channels/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
9.
J Cell Sci ; 112 ( Pt 14): 2397-407, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381395

ABSTRACT

Abnormal proliferation signals, driven by cellular or viral oncogenes, can result in the induction of apoptosis under sub-optimal cell growth conditions. The tumor suppressor p53 plays a central role in mediating oncogene-induced apoptosis, therefore transformed cells lacking p53 are generally resistant to apoptosis-promoting treatments. In a previous work we have reported that the expression of polyomavirus large T antigen causes apoptosis in differentiating myoblasts and that this phenomenon is dependent on the onset of muscle differentiation in the absence of a correct cell cycle arrest. Here we report that polyomavirus large T increases the levels and activity of p53, but these alterations are not involved in the apoptotic mechanism. Apoptosis in polyomavirus large T-expressing myoblasts is not prevented by the expression of a p53 dominant-negative mutant nor it is increased by p53 over-expression. Moreover, forced differentiation induced through the over-expression of the muscle regulatory factor MyoD, leads to apoptosis without altering p53 function and, more significantly, even in a p53-null background. Our results indicate that apoptosis induced by the activation of muscle differentiation pathways in oncogene-expressing cells can occur in a p53-independent manner.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Apoptosis/genetics , Apoptosis/physiology , Genes, p53 , Muscles/cytology , Muscles/virology , Animals , Cell Differentiation , Cell Division , Clone Cells , Gene Expression , Mice , Mice, Knockout , Mutation , MyoD Protein/genetics , MyoD Protein/physiology , Oncogenes , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...