Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
EXCLI J ; 21: 436-453, 2022.
Article in English | MEDLINE | ID: mdl-35391917

ABSTRACT

IKBKE have been associated with numerous cancers. As a result, IKBKE have emerged as potential target for cancer therapy. Accumulating evidence support that IKBKE orchestrate tumor cell survival in cancers. Here we evaluated the possible link between IKBKE and ERK phosphorylation. The effects of IKBKE silencing on MAPK activation in tumor vs. normal cells were evaluated via WB and RT-PCR. Ectopically expressed IKBKE, TPL2 or MEK1 constructs were used to examine the possible interactions among them via co-IP. In vitro kinase assays were performed to understand nature of the observed interactions. In tumors, IKBKE regulates MEK/ERK constitutive activations in vitro and in vivo. IKBKE and TPL2 physically interact and this interaction leads to TPL2 phosphorylation. We describe here a novel regulatory link between IKBKE and constitutive ERK1/2 activation in tumor cells. This new circuitry may be relevant for tumor cell survival in various malignancies.

3.
Gene ; 753: 144796, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32450203

ABSTRACT

Colorectal cancer (CRC) is one of the most common types of cancer which affects the colon and the rectum. Approximately one third of annual CRC mortality occurs due to the late detection of this type of cancer. Therefore, there is an urgent need for more powerful diagnostic and prognostic tools for identification and treatment of colorectal tumorigenesis. Non-coding RNAs (ncRNAs) have been implicated in the pathology of CRC and also linked to metastasis, proliferation, differentiation, migration, angiogenesis and apoptosis in numerous cancers. Recently, attention has turned towards ncRNAs as specific targets for diagnosis, prognosis and treatment of various types of cancers, including CRC. In this review, we have tried to outline the roles of ncRNAs, and their involvement in signaling pathways responsible for the progression of CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Apoptosis/genetics , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Neoplasm/genetics , RNA, Untranslated , Signal Transduction/genetics
4.
FEBS J ; 285(12): 2161-2181, 2018 06.
Article in English | MEDLINE | ID: mdl-29582565

ABSTRACT

Nuclear factor κB (NF-κB) transcription factors are the central hubs of signaling pathways connecting proinflammatory signals to cell survival, proliferation and cytokine production. In cancers, NF-κB signaling influences many aspects of tumor development, from initiation to metastasis. These functions are mediated by tumor-induced plasticity that allows tumor cells to adapt and survive in changing conditions within the tumor microenvironment. Tumor cell plasticity is shaped by the inflammatory microenvironment in tumors. This review focuses on inhibitor of NF-κB kinases, the direct upstream elements of NF-κB regulation, specifically on their conventional and non-conventional functions in animal models of tumorigenesis from the recent literature.


Subject(s)
Gene Expression Regulation, Neoplastic , I-kappa B Kinase/genetics , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Tumor Microenvironment/genetics , Animals , Carcinogenesis , Epithelial-Mesenchymal Transition/genetics , Gene Regulatory Networks , Humans , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , Protein Subunits/genetics , Protein Subunits/metabolism , Signal Transduction
5.
Turk J Biol ; 42(4): 268-278, 2018.
Article in English | MEDLINE | ID: mdl-30814890

ABSTRACT

IKBKE (IKKε) has emerged as a key modulator of multiple substrates, controlling oncogenic pathways in various malignancies. mTOR signaling, required for cellular growth, proliferation, and vascular angiogenesis in cancer, is potentially one of the pathways regulated by IKKε. Upon activation by various stimuli, PI3K/AKT or similar effectors can relieve the inhibitory effect of the TSC1/TSC2 complex through their phosphorylation to favor mTOR/S6K activation in the downstream. Therefore, any activity that interferes with PI3K/AKT or their downstream targets, such as TSC1/2 or GSK3α/ß, may activate the mTOR/S6K pathway for oncogenic transformation in normal cells. Previous studies have shown that PI3K/AKT can be directly phosphoregulated by IKKε. Here, we propose a new regulatory function for IKKε in the mTOR/S6K pathway through its direct interaction with TSC1, leading to TSC1 phosphorylation, which is vital to suppress its inhibitory role in mTOR activation. Experimentally, upon IKKε deficiency in colorectal cancer cells, we observed that S6K activity was diminished while TSC1 levels were found to be stabilized. We hypothesized that these observations may result from direct interaction between IKKε and TSC1. Indeed, the interaction of these two proteins involves the phosphoregulation of TSC1 in various cell lines. Therefore, we propose a mechanism where IKKε, through regulating TSC1 stability in cancer cells, may create an alternative regulatory loop for the activation of mTOR signaling. These results can potentially be important for the development of novel therapeutic strategies targeting mTOR signaling.

7.
Cancer Res ; 76(9): 2587-99, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26980769

ABSTRACT

Constitutive Wnt signaling promotes intestinal cell proliferation, but signals from the tumor microenvironment are also required to support cancer development. The role that signaling proteins play to establish a tumor microenvironment has not been extensively studied. Therefore, we assessed the role of the proinflammatory Ikk-related kinase Ikkε in Wnt-driven tumor development. We found that Ikkε was activated in intestinal tumors forming upon loss of the tumor suppressor Apc Genetic ablation of Ikkε in ß-catenin-driven models of intestinal cancer reduced tumor incidence and consequently extended survival. Mechanistically, we attributed the tumor-promoting effects of Ikkε to limited TNF-dependent apoptosis in transformed intestinal epithelial cells. In addition, Ikkε was also required for lipopolysaccharide (LPS) and IL17A-induced activation of Akt, Mek1/2, Erk1/2, and Msk1. Accordingly, genes encoding pro-inflammatory cytokines, chemokines, and anti-microbial peptides were downregulated in Ikkε-deficient tissues, subsequently affecting the recruitment of tumor-associated macrophages and IL17A synthesis. Further studies revealed that IL17A synergized with commensal bacteria to trigger Ikkε phosphorylation in transformed intestinal epithelial cells, establishing a positive feedback loop to support tumor development. Therefore, TNF, LPS, and IL17A-dependent signaling pathways converge on Ikkε to promote cell survival and to establish an inflammatory tumor microenvironment in the intestine upon constitutive Wnt activation. Cancer Res; 76(9); 2587-99. ©2016 AACR.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-17/metabolism , Intestinal Neoplasms/pathology , Lipopolysaccharides/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Humans , Immunoprecipitation , In Situ Hybridization , Intestinal Neoplasms/metabolism , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Tumor Microenvironment/physiology
8.
J Exp Med ; 212(12): 2057-75, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26527802

ABSTRACT

Tumor initiation in the intestine can rapidly occur from Lgr5(+) crypt columnar stem cells. Dclk1 is a marker of differentiated Tuft cells and, when coexpressed with Lgr5, also marks intestinal cancer stem cells. Here, we show that Elp3, the catalytic subunit of the Elongator complex, is required for Wnt-driven intestinal tumor initiation and radiation-induced regeneration by maintaining a subpool of Lgr5(+)/Dclk1(+)/Sox9(+) cells. Elp3 deficiency dramatically delayed tumor appearance in Apc-mutated intestinal epithelia and greatly prolonged mice survival without affecting the normal epithelium. Specific ablation of Elp3 in Lgr5(+) cells resulted in marked reduction of polyp formation upon Apc inactivation, in part due to a decreased number of Lgr5(+)/Dclk1(+)/Sox9(+) cells. Mechanistically, Elp3 is induced by Wnt signaling and promotes Sox9 translation, which is needed to maintain the subpool of Lgr5(+)/Dclk1(+) cancer stem cells. Consequently, Elp3 or Sox9 depletion led to similar defects in Dclk1(+) cancer stem cells in ex vivo organoids. Finally, Elp3 deficiency strongly impaired radiation-induced intestinal regeneration, in part because of decreased Sox9 protein levels. Together, our data demonstrate the crucial role of Elp3 in maintaining a subpopulation of Lgr5-derived and Sox9-expressing cells needed to trigger Wnt-driven tumor initiation in the intestine.


Subject(s)
Histone Acetyltransferases/metabolism , Intestines/physiopathology , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Regeneration/physiology , Wnt Proteins/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Doublecortin-Like Kinases , Gene Expression/radiation effects , HCT116 Cells , HEK293 Cells , HT29 Cells , Histone Acetyltransferases/genetics , Humans , Intestinal Mucosa/metabolism , Intestines/radiation effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/pathology , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Regeneration/genetics , Regeneration/radiation effects , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Wnt Proteins/genetics
9.
J Immunol ; 194(8): 3970-83, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25780039

ABSTRACT

Endosomes have important roles in intracellular signal transduction as a sorting platform. Signaling cascades from TLR engagement to IRF3-dependent gene transcription rely on endosomes, yet the proteins that specifically recruit IRF3-activating molecules to them are poorly defined. We show that adaptor protein containing a pleckstrin-homology domain, a phosphotyrosine-binding domain, and a leucine zipper motif (APPL)1, an early endosomal protein, is required for both TRIF- and retinoic acid-inducible gene 1-dependent signaling cascades to induce IRF3 activation. APPL1, but not early endosome Ag 1, deficiency impairs IRF3 target gene expression upon engagement of both TLR3 and TLR4 pathways, as well as in H1N1-infected macrophages. The IRF3-phosphorylating kinases TBK1 and IKKε are recruited to APPL1 endosomes in LPS-stimulated macrophages. Interestingly, APPL1 undergoes proteasome-mediated degradation through ERK1/2 to turn off signaling. APPL1 degradation is blocked when signaling through the endosome is inhibited by chloroquine or dynasore. Therefore, APPL1 endosomes are critical for IRF3-dependent gene expression in response to some viral and bacterial infections in macrophages. Those signaling pathways involve the signal-induced degradation of APPL1 to prevent aberrant IRF3-dependent gene expression linked to immune diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , I-kappa B Kinase/immunology , Protein Serine-Threonine Kinases/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Antirheumatic Agents/pharmacology , Chloroquine/pharmacology , Endosomes/genetics , Endosomes/immunology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Hydrazones/pharmacology , I-kappa B Kinase/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Protein Serine-Threonine Kinases/genetics , Proteolysis/drug effects , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics
10.
Nat Commun ; 5: 5232, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25366117

ABSTRACT

Constitutive activation of EGFR- and NF-κB-dependent pathways is a hallmark of cancer, yet signalling proteins that connect both oncogenic cascades are poorly characterized. Here we define KIAA1199 as a BCL-3- and p65-dependent gene in transformed keratinocytes. KIAA1199 expression is enhanced on human papillomavirus (HPV) infection and is aberrantly expressed in clinical cases of cervical (pre)neoplastic lesions. Mechanistically, KIAA1199 binds Plexin A2 and protects from Semaphorin 3A-mediated cell death by promoting EGFR stability and signalling. Moreover, KIAA1199 is an EGFR-binding protein and KIAA1199 deficiency impairs EGF-dependent Src, MEK1 and ERK1/2 phosphorylations. Therefore, EGFR stability and signalling to downstream kinases requires KIAA1199. As such, KIAA1199 promotes EGF-mediated epithelial-mesenchymal transition (EMT). Taken together, our data define KIAA1199 as an oncogenic protein induced by HPV infection and constitutive NF-κB activity that transmits pro-survival and invasive signals through EGFR signalling.


Subject(s)
ErbB Receptors/metabolism , Papillomavirus Infections/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , B-Cell Lymphoma 3 Protein , Cell Survival , Epidermal Growth Factor/metabolism , Epithelial-Mesenchymal Transition , HeLa Cells , Humans , Hyaluronoglucosaminidase , Keratinocytes/metabolism , Lysosomes/metabolism , MCF-7 Cells , Proteins/genetics , Semaphorin-3A/metabolism , Uterine Cervical Dysplasia/metabolism
11.
Cell Rep ; 7(6): 1914-25, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24882009

ABSTRACT

The recruitment of immune cells into solid tumors is an essential prerequisite of tumor development. Depending on the prevailing polarization profile of these infiltrating leucocytes, tumorigenesis is either promoted or blocked. Here, we identify IκB kinase α (IKKα) as a central regulator of a tumoricidal microenvironment during intestinal carcinogenesis. Mice deficient in IKKα kinase activity are largely protected from intestinal tumor development that is dependent on the enhanced recruitment of interferon γ (IFNγ)-expressing M1-like myeloid cells. In IKKα mutant mice, M1-like polarization is not controlled in a cell-autonomous manner but, rather, depends on the interplay of both IKKα mutant tumor epithelia and immune cells. Because therapies aiming at the tumor microenvironment rather than directly at the mutated cancer cell may circumvent resistance development, we suggest IKKα as a promising target for colorectal cancer (CRC) therapy.


Subject(s)
Carcinogenesis/metabolism , I-kappa B Kinase/metabolism , Intestines/immunology , Killer Cells, Natural/pathology , Myeloid Cells/cytology , Myeloid Cells/enzymology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/pathology , Carcinogenesis/pathology , Cell Polarity , Cell Transformation, Neoplastic , HEK293 Cells , Humans , Killer Cells, Natural/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Phosphorylation , Signal Transduction
12.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23273993

ABSTRACT

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Subject(s)
Cell Dedifferentiation , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Colon/pathology , Epithelial Cells/pathology , Female , Humans , Male , Mice , NF-kappa B/metabolism , Wnt Signaling Pathway
13.
Cell ; 130(5): 918-31, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17803913

ABSTRACT

IKKbeta-dependent NF-kappaB activation plays a key role in innate immunity and inflammation, and inhibition of IKKbeta has been considered as a likely anti-inflammatory therapy. Surprisingly, however, mice with a targeted IKKbeta deletion in myeloid cells are more susceptible to endotoxin-induced shock than control mice. Increased endotoxin susceptibility is associated with elevated plasma IL-1beta as a result of increased pro-IL-1beta processing, which was also seen upon bacterial infection. In macrophages enhanced pro-IL-1beta processing depends on caspase-1, whose activation is inhibited by NF-kappaB-dependent gene products. In neutrophils, however, IL-1beta secretion is caspase-1 independent and depends on serine proteases, whose activity is also inhibited by NF-kappaB gene products. Prolonged pharmacologic inhibition of IKKbeta also augments IL-1beta secretion upon endotoxin challenge. These results unravel an unanticipated role for IKKbeta-dependent NF-kappaB signaling in the negative control of IL-1beta production and highlight potential complications of long-term IKKbeta inhibition.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Shock, Septic/metabolism , Animals , Apoptosis , Carbolines/pharmacology , Caspase 1/metabolism , Cell Line , Disease Models, Animal , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Lipopolysaccharides , Macrophages/drug effects , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptors, Interleukin-1/metabolism , Serine Endopeptidases/metabolism , Serpins/metabolism , Shock, Septic/chemically induced , Shock, Septic/enzymology , Shock, Septic/immunology , Shock, Septic/pathology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL