Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Anticancer Res ; 44(5): 1885-1894, 2024 May.
Article in English | MEDLINE | ID: mdl-38677721

ABSTRACT

BACKGROUND/AIM: Breast cancer is a leading cause of cancer-related deaths among women. Down-regulation of the tumor suppressor gene Cyld in breast cancer has been linked to a poor prognosis. This study investigated the role of Cyld in breast cancer using conditional mutant mouse models carrying a Cyld mutation, which inactivates the deubiquitinating activity of its protein product CYLD in mammary epithelial cells. MATERIALS AND METHODS: We examined the potential of CYLD inactivation to induce mammary tumors spontaneously or modify the susceptibility of mice to mammary tumorigenesis by DMBA treatment or ErbB2 over-expression. RESULTS: CYLD inactivation significantly increased susceptibility to breast cancer induced by either DMBA treatment or ErbB2 over-expression. Moreover, while CYLD inactivation alone did not lead to spontaneous mammary tumorigenesis, it did contribute to the formation of multifocal hyperplastic lesions in virgin mice of predominantly FVB/NJ background. CONCLUSION: Our study demonstrates the tumor enhancing potential of CYLD inactivation in mammary tumorigenesis in vivo and establishes novel relevant mouse models that can be exploited for developing prognostic and therapeutic protocols.


Subject(s)
Deubiquitinating Enzyme CYLD , Animals , Female , Mice , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/genetics , Mutation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
2.
Cancers (Basel) ; 15(20)2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37894364

ABSTRACT

CYLD is a tumor suppressor gene coding for a deubiquitinating enzyme that has a critical regulatory function in a variety of signaling pathways and biological processes involved in cancer development and progression, many of which are also key modulators of somatic cell reprogramming. Nevertheless, the potential role of CYLD in this process has not been studied. With the dual aim of investigating the involvement of CYLD in reprogramming and developing a better understanding of the intricate regulatory system governing this process, we reprogrammed control (CYLDWT/WT) and CYLD DUB-deficient (CYLDΔ9/Δ9) mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells (iPSCs) through ectopic overexpression of the Yamanaka factors (Oct3/4, Sox2, Klf4, c-myc). CYLD DUB deficiency led to significantly reduced reprogramming efficiency and slower early reprogramming kinetics. The introduction of WT CYLD to CYLDΔ9/Δ9 MEFs rescued the phenotype. Nevertheless, CYLD DUB-deficient cells were capable of establishing induced pluripotent colonies with full spontaneous differentiation potential of the three germ layers. Whole proteome analysis (Data are available via ProteomeXchange with identifier PXD044220) revealed that the mesenchymal-to-epithelial transition (MET) during the early reprogramming stages was disrupted in CYLDΔ9/Δ9 MEFs. Interestingly, differentially enriched pathways revealed that the primary processes affected by CYLD DUB deficiency were associated with the organization of the extracellular matrix and several metabolic pathways. Our findings not only establish for the first time CYLD's significance as a regulatory component of early reprogramming but also highlight its role as an extracellular matrix regulator, which has profound implications in cancer research.

3.
Cancers (Basel) ; 12(8)2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32722292

ABSTRACT

Downregulation of the cylindromatosis (CYLD) tumor suppressor has been associated with breast cancer development and progression. Here, we report a critical role for CYLD in maintaining the phenotype of mammary epithelial cells in vitro and in vivo. CYLD downregulation or inactivation induced an epithelial to mesenchymal transition of mammary epithelial cells that was dependent on the concomitant activation of the transcription factors Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) and transforming growth factor beta (TGF)signaling. CYLD inactivation enhanced the nuclear localization of YAP/TAZ and the phosphorylation of Small Mothers Against Decapentaplegic (SMAD)2/3 proteins in confluent cell culture conditions. Consistent with these findings were the hyperplastic alterations of CYLD-deficient mouse mammary epithelia, which were associated with enhanced nuclear expression of the YAP/TAZ transcription factors. Furthermore, in human breast cancer samples, downregulation of CYLD expression correlates with enhanced YAP/TAZ-regulated target gene expression. Our results identify CYLD as a critical regulator of a signaling node that prevents the coordinated activation of YAP/TAZ and the TGF pathway in mammary epithelial cells, in order to maintain their phenotypic identity and homeostasis. Consequently, they provide a novel conceptual framework that supports and explains a causal implication of deficient CYLD expression in aggressive human breast cancers.

4.
Onco Targets Ther ; 13: 5575-5588, 2020.
Article in English | MEDLINE | ID: mdl-32606772

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a frequently diagnosed cancer and a leading cause of cancer-related death worldwide. Its rapid progression, combined with the limited treatment options at late stages, imposes the need for early detection and aggressive intervention. Based on the knowledge that hepatocarcinogenesis is significantly influenced by histone acetylation, we directed our search for novel HCC therapeutics among histone deacetylation inhibitors (HDACi). The aim of the present study was to investigate the effect of HDAC1/2 inhibitor Romidepsin in the well-established mouse model of diethylnitrosamine (DEN)-induced HCC. MATERIALS AND METHODS: C56BL/6 mice were treated with Romidepsin at the critical point of 10 months after DEN challenge and their livers were examined 2 months later using histopathology and morphometry. Protein levels were assessed in serum using ELISA and in liver tissues using Western blot and immunohistochemistry (in-situ detection). Gene expression was quantified using real-time PCR. RESULTS: Romidepsin suppressed cancer progression. This effect was associated with decreased proliferation and increased apoptosis of cancer cells. The cell cycle regulator CK2a, the anti-inflammatory molecule PPAR-γ, and the tumor suppressors PTEN and CYLD were upregulated in treated HCC. By contrast, the expression of PI3K, NF-κB p65 and c-Jun was reduced. In line with this result, the levels of two major apoptosis regulators, ie, BAD and the multifunctional protein c-Met, were lower in the blood serum of treated mice compared to the untreated mice with HCC. CONCLUSION: These findings suggest that Romidepsin, a drug currently used in the treatment of lymphoma, could also be considered in the management of early-stage HCC.

5.
Nat Immunol ; 21(6): 626-635, 2020 06.
Article in English | MEDLINE | ID: mdl-32424362

ABSTRACT

The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Enterocolitis/etiology , Enterocolitis/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptors, Cell Surface/metabolism , Animals , Citrobacter rodentium , Deubiquitinating Enzyme CYLD/genetics , Disease Models, Animal , Disease Susceptibility , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Enterocolitis/pathology , Gene Expression , Humans , Interleukin-18/antagonists & inhibitors , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Protein Binding/immunology , Ubiquitination
6.
PLoS One ; 13(2): e0191864, 2018.
Article in English | MEDLINE | ID: mdl-29394249

ABSTRACT

The human cylindromatosis tumor suppressor (HsCyld) has attracted extensive attention due to its association with the development of multiple types of cancer. HsCyld encodes a deubiquitinating enzyme (HsCYLD) with a broad range of functions that include the regulation of several cell growth, differentiation and death pathways. HsCyld is an evolutionarily conserved gene. Homologs of HsCyld have been identified in simple model organisms such as Drosophila melanogaster and Caenorhabditis elegans (C. elegans) which offer extensive possibilities for functional analyses. In the present report we have investigated and compared the functional properties of HsCYLD and its C. elegans homolog (CeCYLD). As expected from the mammalian CYLD expression pattern, the CeCyld promoter is active in multiple tissues with certain gastrointestinal epithelia and neuronal cells showing the most prominent activity. CeCYLD is a functional deubiquitinating enzyme with similar specificity to HsCYLD towards K63- and M1-linked polyubiquiting chains. CeCYLD was capable of suppressing the TRAF2-mediated activation of NF-kappaB and AP1 similarly to HsCYLD. Finally, CeCYLD could suppress the induction of TNF-dependent gene expression in mammalian cells similarly to HsCYLD. Our results demonstrate extensively overlapping functions between the HsCYLD and CeCYLD, which establish the C. elegans protein as a valuable model for the elucidation of the complex activity of the human tumor suppressor protein.


Subject(s)
Caenorhabditis elegans/genetics , Genes, Helminth , Amino Acid Sequence , Animals , Humans , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid
7.
Anticancer Res ; 37(7): 3493-3503, 2017 07.
Article in English | MEDLINE | ID: mdl-28668838

ABSTRACT

BACKGROUND/AIM: The cylindromatosis tumor suppressor (CYLD) has been implicated in the inhibition of human breast cancer development by virtue of the poor prognosis of patients with down-regulated CYLD expression. In order to investigate the mechanism of breast cancer suppression by CYLD, in the present study, cellular and molecular aspects of CYLD-dependent phenotypic regulation of different types of human breast cancer cell lines were analyzed. MATERIALS AND METHODS: CYLD expression was down-regulated by RNA interference in human breast cancer cell lines. Parental and CYLD-deficient cell lines were evaluated for their viability, migratory capacity, anchorage-independent growth and chemoresistance. Wild-type and mutated forms of CYLD were also evaluated for their ability to suppress the clonogenic potential of breast cancer cells. RESULTS: CYLD down-regulation enhanced the survival and migratory properties of basal and luminal breast cancer cell lines. In addition, down-regulation of CYLD expression enhanced the ability of human breast cancer cells to grow in an anchorage-independent manner and could be associated with resistance to chemotherapeutic drugs. The growth-suppressive properties of CYLD on breast cancer cell lines were dependent on its de-ubiquitinating activity and its amino terminal cytoskeleton-interacting region. CONCLUSION: Our results establish a broad range of tumor-suppressive properties that are conferred by CYLD in basal and luminal human breast cancer cells and support the significance of targeted de-ubiquitination by CYLD in breast cancer cell growth suppression.


Subject(s)
Breast Neoplasms/genetics , Down-Regulation/genetics , Genes, Tumor Suppressor/physiology , Tumor Suppressor Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Cytoskeleton/genetics , Deubiquitinating Enzyme CYLD , Drug Resistance, Neoplasm/genetics , Female , HEK293 Cells , Humans , MCF-7 Cells , RNA Interference/physiology , Ubiquitination/genetics
8.
Int J Biochem Cell Biol ; 82: 49-56, 2017 01.
Article in English | MEDLINE | ID: mdl-27865894

ABSTRACT

Several studies have implicated the downregulation of the tumor suppressor Cyld expression in breast cancer development. However, the mechanisms that regulate Cyld expression in mammary epithelial cells are largely unknown. In order to investigate them, a bioinformatic analysis of the promoter region of Cyld was performed and identified putative nuclear hormone receptor response elements that included peroxisome proliferator-activated receptor gamma (PPAR-γ)-responsive elements. In the present study, we showed that upon activation of the nuclear hormone receptor PPAR-γ by the agonist troglitazone (TZD), there was a significant increase in Cyld mRNA in human mammary epithelial cell lines. The effect of TZD could be attributed to the transactivation of the Cyld promoter as indicated by the upregulation of a luciferase reporter that was driven by the -1995 to +95 region of the human Cyld gene. Furthermore, the upregulation of Cyld expression by TZD was dependent on PPAR-γ since downregulation of PPAR-γ expression by RNAi compromised the induction of Cyld expression by TZD. CYLD induction mediated, at least in part, the TZD-mediated downregulation of tumor necrosis factor α (TNFα)-induced interleukin 8 (IL-8). In addition, downregulation of CYLD compromised the cytotoxic effects of TZD in immortalized mammary epithelial cells. Our results demonstrated that PPAR-γ is a novel regulator of Cyld transcription and identified CYLD as a mediator of the PPAR-γ-dependent anti-inflammatory and anti-proliferative activity in mammary epithelial cells, which underscores its potential to be used as a target for the development of breast cancer therapeutic approaches.


Subject(s)
Breast Neoplasms/drug therapy , Chromans/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hypoglycemic Agents/pharmacology , Mammary Glands, Human/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Tumor Suppressor Proteins/agonists , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Computational Biology , Deubiquitinating Enzyme CYLD , Female , Gene Expression Regulation/drug effects , Humans , Mammary Glands, Human/immunology , Mammary Glands, Human/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic/drug effects , RNA Interference , RNA, Messenger/metabolism , Response Elements/drug effects , Troglitazone , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Cell Biochem Funct ; 34(7): 465-468, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27570065

ABSTRACT

CYLD is a deubiquitinating enzyme that exerts a tumor suppressive function. Its downregulation or inactivation has been associated with the development of several types of malignancies including hepatocellular carcinoma (HCC). HCC cells display significantly lower Cyld expression compared to primary human hepatocytes, and Cyld downregulation can contribute to apoptotic resistance of HCC cells. Little is known about the mechanism of Cyld downregulation in human HCC cells. In the present study we explored the possible regulation of Cyld expression by histone deacetylases (HDACs) in human HCC cell lines. We demonstrated that the HDAC inhibitors suberoylanilide hydroxamic acid, sodium butyrate, and trichostatin A induced the upregulation of both mRNA and protein levels of CYLD in two different HCC cell lines, HepG2 and Huh7. Our results demonstrate the involvement of HDACs in the downregulation of Cyld expression in HCC cells and support and may improve the use of HDAC inhibitors for the treatment for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Histone Deacetylase Inhibitors/pharmacology , Liver Neoplasms/genetics , Up-Regulation/drug effects , Butyric Acid/pharmacology , Cell Line, Tumor , Deubiquitinating Enzyme CYLD , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vorinostat
10.
JCI Insight ; 1(11)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27478875

ABSTRACT

The deubiquitinase-encoding gene Cyld displays a dominant genetic linkage to a wide spectrum of skin-appendage tumors, which could be collectively designated as CYLD mutant-syndrome (CYLDm-syndrome). Despite recent advances, little is understood about the molecular mechanisms responsible for this painful and difficult-to-treat skin disease. Here, we generated a conditional mouse model with epidermis-targeted expression of a catalytically deficient CYLDm through K14-Cre-mediated deletion of exon 9 (hereafter refer to CyldEΔ9/Δ9 ). CyldEΔ9/Δ9 mice were born alive but developed hair and sebaceous gland abnormalities and dental defects at 100% and 60% penetrance, respectively. Upon topical challenge with DMBA/TPA, these animals primarily developed sebaceous and basaloid tumors resembling human CYLDm-syndrome as opposed to papilloma, which is most commonly induced in WT mice by this treatment. Molecular analysis revealed that TRAF6-K63-Ubiquitination (K63-Ub), c-Myc-K63-Ub, and phospho-c-Myc (S62) were markedly elevated in CyldEΔ9/Δ9 skin. Topical treatment with a pharmacological c-Myc inhibitor induced sebaceous and basal cell apoptosis in CyldEΔ9/Δ9 skin. Consistently, c-Myc activation was readily detected in human cylindroma and sebaceous adenoma. Taken together, our findings demonstrate that CyldEΔ9/Δ9 mice represent a disease-relevant animal model and identify TRAF6 and c-Myc as potential therapeutic targets for CYLDm-syndrome.

11.
Cell Oncol (Dordr) ; 39(3): 287-93, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27042826

ABSTRACT

PURPOSE: CYLD is a tumor suppressor that has been linked to the development of various human malignancies, including colon cancer. The tumor-suppressing function of CYLD is associated with its deubiquitinating activity, which maps to the carboxyl-terminal region of the protein. In the present study we evaluated the role of intestinal epithelial CYLD in colitis-associated cancer using a conditional mouse CYLD inactivation model. METHODS: In order to evaluate the role of CYLD in intestinal epithelial carcinogenesis, mice (IEC-Cyld (Δ9) mice) that carry a mutation that eliminates the deubiquitinating domain of CYLD in intestinal epithelial cells (IEC) were generated by crossing Villin-Cre transgenic mice to previously generated mice carrying a loxP-flanked Cyld exon 9 (Cyld (flx9) mice). RESULTS: We found that IEC-Cyld (Δ9) mice did not present spontaneous intestinal abnormalities up to one year of age. However, upon challenge with a combination of genotoxic (AOM) and pro-inflammatory (DSS) agents we found that the number of adenomas in the IEC-Cyld (Δ9) mice was dramatically increased compared to the control mice. Inactivation of CYLD in intestinal epithelial cells did not affect the classical nuclear factor-kappaB (NF-κB) and c-Jun kinase (JNK) activation pathways under physiological conditions, suggesting that these pathways do not predispose CYLD-deficient intestinal epithelia to colorectal cancer development before the onset of genotoxic and/or pro-inflammatory stress. CONCLUSIONS: Our findings underscore a critical tumor-suppressing role for functional intestinal epithelial CYLD in colitis-associated carcinogenesis. CYLD expression and its associated pathways in intestinal tumors may be exploited for future prognostic and therapeutic purposes.


Subject(s)
Carcinogenesis/genetics , Colitis/complications , Colorectal Neoplasms/genetics , Cysteine Endopeptidases/genetics , Intestinal Mucosa/pathology , Animals , Colitis/genetics , Deubiquitinating Enzyme CYLD , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Cell Signal ; 26(12): 2730-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25134449

ABSTRACT

TRAFs constitute a family of proteins that have been implicated in signal transduction by immunomodulatory cellular receptors and viral proteins. TRAF2 and TRAF6 have an E3-ubiquitin ligase activity, which is dependent on the integrity of their RING finger domain and it has been associated with their ability to activate the NF-κB and AP1 signaling pathways. A yeast two-hybrid screen with TRAF2 as bait, identified the regulatory subunit PP4R1 of protein phosphatase PP4 as a TRAF2-interacting protein. The interaction of TRAF2 with PP4R1 depended on the integrity of the RING finger domain of TRAF2. PP4R1 could interact also with the TRAF2-related factor TRAF6 in a RING domain-dependent manner. Exogenous expression of PP4R1 inhibited NF-κB activation by TRAF2, TRAF6, TNF and the Epstein-Barr virus oncoprotein LMP1. In addition, expression of PP4R1 downregulated IL8 induction by LMP1, whereas downregulation of PP4R1 by RNA interference enhanced the induction of IL8 by LMP1 and TNF. PP4R1 could mediate the dephosphorylation of TRAF2 Ser11, which has been previously implicated in TRAF2-mediated activation of NF-κB. Finally, PP4R1 could inhibit TRAF6 polyubiquitination, suggesting an interference with the E3 ubiquitin ligase activity of TRAF6. Taken together, our data identify a novel mechanism of NF-κB pathway inhibition which is mediated by PP4R1-dependent targeting of specific TRAF molecules.


Subject(s)
NF-kappa B/metabolism , Phosphoprotein Phosphatases/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/metabolism , Down-Regulation/physiology , HEK293 Cells , Humans , Interleukin-8/metabolism , RING Finger Domains/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Viral Matrix Proteins
13.
PLoS One ; 9(4): e95688, 2014.
Article in English | MEDLINE | ID: mdl-24759913

ABSTRACT

Epstein-Barr virus (EBV) is a human herpesvirus, which is causally associated with the development of several B lymphocytic malignancies that include Burkitt's lymphomas, Hodgkin's disease, AIDS and posttransplant associated lymphomas. The transforming activity of EBV is orchestrated by several latent viral proteins that mimic and modulate cellular growth promoting and antiapoptotic signaling pathways, which involve among others the activity of protein kinases. In an effort to identify small molecule inhibitors of the growth of EBV-transformed B lymphocytes a library of 254 kinase inhibitors was screened. This effort identified two tyrosine kinase inhibitors and two MEK inhibitors that compromised preferentially the viability of EBV-infected human B lymphocytes. Our findings highlight the possible dependence of EBV-infected B lymphocytes on specific kinase-regulated pathways underlining the potential for the development of small molecule-based therapeutics that could target selectively EBV-associated human B lymphocyte malignancies.


Subject(s)
B-Lymphocytes/virology , Cell Survival/drug effects , Herpesvirus 4, Human/pathogenicity , Protein Kinase Inhibitors/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Line , Humans
14.
Proc Natl Acad Sci U S A ; 109(51): 21022-7, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23223530

ABSTRACT

Germinal centers (GCs) are specialized microenvironments in secondary lymphoid organs where high-affinity antibody-producing B cells are selected based on B-cell antigen receptor (BCR) signal strength. BCR signaling required for normal GC selection is uncertain. We have found that protein kinase N1 (PKN1, also known as PRK1) negatively regulates Akt kinase downstream of the BCR and that this regulation is necessary for normal GC development. PKN1 interacted with and inhibited Akt1 kinase and transforming activities. Pkn1(-/-) B cells were hyperresponsive and had increased phosphorylated Akt1 levels upon BCR stimulation. In the absence of immunization or infection, Pkn1(-/-) mice spontaneously formed GCs and developed an autoimmune-like disease with age, which was characterized by autoantibody production and glomerulonephritis. More B cells, with fewer somatic BCR gene V region hypermutations were selected in Pkn1(-/-) GCs. These results indicate that PKN1 down-regulation of BCR-activated Akt activity is critical for normal GC B-cell survival and selection.


Subject(s)
Gene Expression Regulation, Enzymologic , Germinal Center/immunology , Protein Kinase C/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Autoantibodies/chemistry , B-Lymphocytes/metabolism , Cell Proliferation , Down-Regulation , Glomerulonephritis/metabolism , Immunity, Humoral , Immunoglobulins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction
15.
Cancer Cell ; 21(6): 738-50, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22698400

ABSTRACT

The tumor suppressor cylindromatosis (CYLD) inhibits the NFκB and mitogen-activated protein kinase (MAPK) activation pathways by deubiquitinating upstream regulatory factors. Here we show that liver-specific disruption of CYLD triggers hepatocyte cell death in the periportal area via spontaneous and chronic activation of TGF-ß activated kinase 1 (TAK1) and c-Jun N-terminal kinase (JNK). This is followed by hepatic stellate cell and Kupffer cell activation, which promotes progressive fibrosis, inflammation, tumor necrosis factor (TNF) production, and expansion of hepatocyte apoptosis toward the central veins. At later stages, compensatory proliferation results in the development of cancer foci featuring re-expression of oncofetal hepatic and stem cell-specific genes. The results demonstrate that, in the liver, CYLD acts as an important regulator of hepatocyte homeostasis, protecting cells from spontaneous apoptosis by preventing uncontrolled TAK1 and JNK activation.


Subject(s)
Apoptosis/genetics , Cysteine Endopeptidases/genetics , Hepatocytes/metabolism , Liver/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Comparative Genomic Hybridization , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Enzyme Activation/drug effects , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Profiling , Hepatocytes/pathology , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
J Biol Chem ; 286(44): 38768-38782, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21896491

ABSTRACT

The transcription factor NF-κB is a critical regulator of immune responses. To determine how NF-κB builds transcriptional control networks, we need to obtain a topographic map of the factor bound to the genome and correlate it with global gene expression. We used a ChIP cloning technique and identified novel NF-κB target genes in response to virus infection. We discovered that most of the NF-κB-bound genomic sites deviate from the consensus and are located away from conventional promoter regions. Remarkably, we identified a novel abundant NF-κB-binding site residing in specialized Alu-repetitive elements having the potential for long range transcription regulation, thus suggesting that in addition to its known role, NF-κB has a primate-specific function and a role in human evolution. By combining these data with global gene expression profiling of virus-infected cells, we found that most of the sites bound by NF-κB in the human genome do not correlate with changes in gene expression of the nearby genes and they do not appear to function in the context of synthetic promoters. These results demonstrate that repetitive elements interspersed in the human genome function as common target sites for transcription factors and may play an important role in expanding the repertoire of binding sites to engage new genes into regulatory networks.


Subject(s)
Alu Elements/genetics , NF-kappa B/metabolism , Animals , Binding Sites , Chromatin/chemistry , Chromatin Immunoprecipitation , DNA/chemistry , DNA/genetics , Genome , Genome, Human , HeLa Cells , Humans , Mice , Oligonucleotide Array Sequence Analysis , Primates , Protein Binding , Transcription, Genetic
17.
Eur J Immunol ; 41(10): 3054-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21728169

ABSTRACT

The cylindromatosis tumor suppressor gene (Cyld) encodes an enzyme (CYLD) with deubiquitinating activity that has been implicated in the regulation of thymocyte selection in an NF-κB-essential-modulator (NEMO)-dependent manner. The main known molecular defects in thymocytes with inactive CYLD (LckCre-Cyld(flx9/flx9) ) are the aberrant hyperactivation of NF-κB and JNK pathways. In order to dissect further the molecular mechanism of CYLD-dependent thymocyte selection and address the role of NF-κB specifically, we generated double mutant mice (LckCre-Cyld(flx9/flx9) -Ikk2(flx/flx) ) in which CYLD was inactivated concomitantly with IKK2 (IκB-kinase 2) in thymocytes. Interestingly, thymic development and NF-κB activity in double mutant mice were fully restored, indicating that an IKK2-dependent function of CYLD that leads to the hyperactivation of the NF-κB pathway is primarily responsible for the defective selection of thymocytes. Intriguingly, we observed a greater reduction of CD4(+) and CD8(+) T cells in the periphery of LckCre-Cyld(flx9/flx9) -Ikk2(flx/flx) mice compared with LckCre-Ikk2(flx/flx) mice. Collectively, our data establish CYLD as a critical regulator of thymocyte selection in a manner that depends on IKK2 and NF-κB activation. In addition, our data uncover an IKK2-independent role for CYLD in the establishment of physiological T-cell populations in the periphery.


Subject(s)
Cysteine Endopeptidases/metabolism , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Electrophoretic Mobility Shift Assay , Flow Cytometry , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Immunoblotting , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thymus Gland/immunology
18.
PLoS One ; 6(1): e16397, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21283724

ABSTRACT

The cylindromatosis tumor suppressor (CYLD) is a deubiquitinating enzyme that has been implicated in various aspects of adaptive and innate immune responses. Nevertheless, the role of CYLD in the function of specific types of immune cells remains elusive. In this report we have used conditional gene targeting in mice to address the role of the deubiquitinating activity of CYLD in the myelomonocytic lineage. Truncation of the deubiquitinating domain of CYLD specifically in myelomonocytic cells impaired the development of lethal LPS-induced endotoxic shock and the accumulation of thioglycollate-elicited peritoneal macrophages. Our data establish CYLD as a regulator of monocyte-macrophage activation in response to inflammatory stimuli and identify it as a potential target for therapeutic intervention in relevant inflammatory disorders in humans.


Subject(s)
Cysteine Endopeptidases , Inflammation/prevention & control , Leukemia, Myelomonocytic, Chronic/enzymology , Ubiquitination , Animals , Deubiquitinating Enzyme CYLD , Immunity , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages, Peritoneal , Mice , Monocytes , Shock, Septic/prevention & control
19.
Cell Signal ; 23(5): 772-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21185369

ABSTRACT

TRAF6 is an E3 ubiquitin ligase that plays a pivotal role in the activation of NF-κB by innate and adaptive immunity stimuli. TRAF6 consists of a highly conserved carboxyl terminal TRAF-C domain which is preceded by a coiled coil domain and an amino terminal region that contains a RING domain and a series of putative zinc-finger motifs. The TRAF-C domain contributes to TRAF6 oligomerization and mediates the interaction of TRAF6 with upstream signaling molecules whereas the RING domain comprises the core of the ubiquitin ligase catalytic domain. In order to identify structural elements that are important for TRAF6-induced NF-κB activation, mutational analysis of the TRAF-C and RING domains was performed. Alterations of highly conserved residues of the TRAF-C domain of TRAF6 did not affect significantly the ability of the protein to activate NF-κB. On the other hand a number of functionally important residues (L77, Q82, R88, F118, N121 and E126) for the activation of NF-κB were identified within the RING domain of TRAF6. Interestingly, several homologues of these residues in TRAF2 were shown to have a conserved functional role in TRAF2-induced NF-κB activation and lie at the dimerization interface of the RING domain. Finally, whereas alteration of Q82, R88 and F118 compromised both the K63-linked polyubiquitination of TRAF6 and its ability to activate NF-κB, alteration of L77, N121 and E126 diminished the NF-κB activating function of TRAF6 without affecting TRAF6 K63-linked polyubiquitination. Our results support a conserved functional role of the TRAF RING domain dimerization interface and a potentially necessary but insufficient role for RING-dependent TRAF6 K63-linked polyubiquitination towards NF-κB activation in cells.


Subject(s)
NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism , Cell Line , Dimerization , Humans , Mutagenesis, Site-Directed , Protein Structure, Tertiary , TNF Receptor-Associated Factor 2/chemistry , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/genetics , Ubiquitination , Zinc Fingers
20.
Biochem Biophys Res Commun ; 403(3-4): 335-9, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-21078302

ABSTRACT

Glutaredoxin-1 (GRX-1) is a cytoplasmic enzyme that highly contributes to the antioxidant defense system. It catalyzes the reversible reduction of glutathione-protein mixed disulfides, a process called deglutathionylation. Here, we investigated the role of GRX-1 in the pathway triggered by interleukin-1/Toll-like receptor 4 (IL-1R/TLR4) by using RNA interference (RNAi) in HEK293 and HeLa cells. TNF receptor-associated factor 6 (TRAF6) is an intermediate signalling molecule involved in the signal transduction by members of the interleukin-1/Toll-like receptor (IL-1R/TLR) family. TRAF6 has an E3 ubiquitin ligase activity which depends on the integrity of an amino-terminal really interesting new gene (RING) finger motif. Upon receptor activation, TRAF6 undergoes K63-linked auto-polyubiquitination which mediates protein-protein interactions and signal propagation. Our data showed that IL-1R and TLR4-mediated NF-κB induction was severely reduced in GRX-1 knockdown cells. We found that the RING-finger motif of TRAF6 is S-glutathionylated under normal conditions. Moreover, upon IL-1 stimulation TRAF6 undergoes deglutathionylation catalyzed by GRX-1. The deglutathionylation of TRAF6 is essential for its auto-polyubiquitination and subsequent activation. Taken together, our findings reveal another signalling molecule affected by S-glutathionylation and uncover a crucial role for GRX-1 in the TRAF6-dependent activation of NF-κB by IL-1R/TLRs.


Subject(s)
Glutaredoxins/metabolism , Glutathione/metabolism , Protein Processing, Post-Translational , Receptors, Interleukin-1/metabolism , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism , HEK293 Cells , HeLa Cells , Humans , NF-kappa B/metabolism , RING Finger Domains , Signal Transduction , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...