Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
In Vitro Cell Dev Biol Anim ; 60(5): 449-465, 2024 May.
Article in English | MEDLINE | ID: mdl-38383910

ABSTRACT

Wnt signaling plays a crucial role in embryonic development and homeostasis maintenance. Delicate and sensitive fine-tuning of Wnt signaling based on the proper timings and positions is required to balance cell proliferation and differentiation and maintain individual health. Therefore, homeostasis is broken by tissue hypoplasia or tumor formation once Wnt signal dysregulation disturbs the balance of cell proliferation. The well-known regulatory mechanism of Wnt signaling is the molecular reaction associated with the cytoplasmic accumulation of effector ß-catenin. In addition to ß-catenin, most Wnt effector proteins are also regulated by ubiquitin-dependent modification, both qualitatively and quantitatively. This review will explain the regulation of the whole Wnt signal in four regulatory phases, as well as the different ubiquitin ligases and the function of deubiquitinating enzymes in each phase. Along with the recent results, the mechanism by which RNF43 negatively regulates the surface expression of Wnt receptors, which has recently been well understood, will be detailed. Many RNF43 mutations have been identified in pancreatic and gastrointestinal cancers and examined for their functional alteration in Wnt signaling. Several mutations facilitate or activate the Wnt signal, reversing the RNF43 tumor suppressor function into an oncogene. RNF43 may simultaneously play different roles in classical multistep tumorigenesis, as both wild-type and mutant RNF43 suppress the p53 pathway. We hope that the knowledge obtained from further research in RNF43 will be applied to cancer treatment in the future despite the fully unclear function of RNF43.


Subject(s)
Carcinogenesis , Receptors, Wnt , Wnt Signaling Pathway , Humans , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Animals , Wnt Signaling Pathway/genetics , Receptors, Wnt/metabolism , Receptors, Wnt/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
2.
J Invest Dermatol ; 142(6): 1576-1586.e2, 2022 06.
Article in English | MEDLINE | ID: mdl-34742703

ABSTRACT

Hemidesmosomes (HDs) are adhesion complexes that promote epithelial-stromal attachment in stratified and complex epithelia, including the epidermis. In various biological processes, such as differentiation and migration of epidermal keratinocytes during wound healing or carcinoma invasion, quick assembly and disassembly of HDs are prerequisites. In this study, we show that inhibition of Wnt/ß-catenin signaling disturbs HD organization in keratinocytes. Screening with inhibitors identified the depletion of HD components and HD-like structures through Wnt inhibition, but keratinocyte differentiation was not affected. Wnt inhibition significantly diminished plectin and type XVII collagen expression in the basal side of Wnt-inhibited cells and the dermo-epidermal junction of the Wnt-inactive murine basal epidermis. Similar to Wnt inhibition, PLEC-knockout cells or cells with plectin-type XVII collagen binding defects showed type XVII collagen reduction in the basal side of the cells, implying the possible involvement of Wnt/ß-catenin signaling in HD assembly. Atypical protein kinase C inhibition ameliorated the phenotypes of Wnt-inhibited cells. These findings show that Wnt/ß-catenin signaling regulates the localization of HD components in keratinocytes and that the atypical protein kinase C pathway is involved in Wnt inhibition‒induced HD disarrangement. Our study suggests that the Wnt signaling pathway could be a potential therapeutic target for treating HD-defective diseases, such as epidermolysis bullosa.


Subject(s)
Hemidesmosomes , beta Catenin , Animals , Hemidesmosomes/metabolism , Keratinocytes/metabolism , Mice , Plectin , Wnt Signaling Pathway , Wound Healing/physiology , beta Catenin/metabolism
3.
Bioessays ; 43(4): e2000297, 2021 04.
Article in English | MEDLINE | ID: mdl-33569855

ABSTRACT

Wnt signaling plays pivotal roles during our entire lives, from conception to death, through the regulation of morphogenesis in developing embryos and the maintenance of tissue homeostasis in adults. The regulation of Wnt signaling occurs on several levels: at the receptor level on the plasma membrane, at the ß-catenin protein level in the cytoplasm, and through transcriptional regulation in the nucleus. Several recent studies have focused on the mechanisms of Wnt receptor regulation, following the discovery that the Wnt receptor frizzled (Fzd) is a target of the ubiquitin ligases, RNF43 and ZNRF3. RNF43 and ZNRF3 are homologous genes that are mutated in several cancers. The details underlying their mechanism of action continue to unfold, while at the same time raising many new questions. In this review, we discuss advances and controversies in our understanding of Wnt receptor regulation.


Subject(s)
Ubiquitin-Protein Ligases , Ubiquitin , Homeostasis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway
4.
Nat Commun ; 11(1): 4586, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934222

ABSTRACT

Frequent mutation of the tumour suppressor RNF43 is observed in many cancers, particularly colon malignancies. RNF43, an E3 ubiquitin ligase, negatively regulates Wnt signalling by inducing degradation of the Wnt receptor Frizzled. In this study, we discover that RNF43 activity requires phosphorylation at a triplet of conserved serines. This phospho-regulation of RNF43 is required for zebrafish development and growth of mouse intestinal organoids. Cancer-associated mutations that abrogate RNF43 phosphorylation cooperate with active Ras to promote tumorigenesis by abolishing the inhibitory function of RNF43 in Wnt signalling while maintaining its inhibitory function in p53 signalling. Our data suggest that RNF43 mutations cooperate with KRAS mutations to promote multi-step tumorigenesis via the Wnt-Ras-p53 axis in human colon cancers. Lastly, phosphomimetic substitutions of the serine trio restored the tumour suppressive activity of extracellular oncogenic mutants. Therefore, harnessing phospho-regulation of RNF43 might be a potential therapeutic strategy for tumours with RNF43 mutations.


Subject(s)
Carcinogenesis/metabolism , Receptors, Wnt/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carcinogenesis/genetics , Humans , Mice , Mice, Inbred BALB C , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Phosphorylation , Proteolysis , Receptors, Wnt/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway
5.
Nat Commun ; 11(1): 1063, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32102997

ABSTRACT

Mediator is a coregulatory complex that regulates transcription of Pol II-dependent genes. Previously, we showed that human Mediator subunit MED26 plays a role in the recruitment of Super Elongation Complex (SEC) or Little Elongation Complex (LEC) to regulate the expression of certain genes. MED26 plays a role in recruiting SEC to protein-coding genes including c-myc and LEC to small nuclear RNA (snRNA) genes. However, how MED26 engages SEC or LEC to regulate distinct genes is unclear. Here, we provide evidence that MED26 recruits LEC to modulate transcription termination of non-polyadenylated transcripts including snRNAs and mRNAs encoding replication-dependent histone (RDH) at Cajal bodies. Our findings indicate that LEC recruited by MED26 promotes efficient transcription termination by Pol II through interaction with CBC-ARS2 and NELF/DSIF, and promotes 3' end processing by enhancing recruitment of Integrator or Heat Labile Factor to snRNA or RDH genes, respectively.


Subject(s)
Gene Expression Regulation/genetics , Mediator Complex/genetics , RNA, Small Nuclear/genetics , Transcription Termination, Genetic/physiology , Transcriptional Elongation Factors/genetics , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/metabolism , RNA Cap-Binding Proteins/metabolism , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
6.
Elife ; 62017 07 11.
Article in English | MEDLINE | ID: mdl-28693719

ABSTRACT

Type XVII collagen (COL17) is a transmembrane protein located at the epidermal basement membrane zone. COL17 deficiency results in premature hair aging phenotypes and in junctional epidermolysis bullosa. Here, we show that COL17 plays a central role in regulating interfollicular epidermis (IFE) proliferation. Loss of COL17 leads to transient IFE hypertrophy in neonatal mice owing to aberrant Wnt signaling. The replenishment of COL17 in the neonatal epidermis of COL17-null mice reverses the proliferative IFE phenotype and the altered Wnt signaling. Physical aging abolishes membranous COL17 in IFE basal cells because of inactive atypical protein kinase C signaling and also induces epidermal hyperproliferation. The overexpression of human COL17 in aged mouse epidermis suppresses IFE hypertrophy. These findings demonstrate that COL17 governs IFE proliferation of neonatal and aged skin in distinct ways. Our study indicates that COL17 could be an important target of anti-aging strategies in the skin.


Subject(s)
Autoantigens/metabolism , Cell Proliferation , Epidermis/physiology , Non-Fibrillar Collagens/metabolism , Animals , Autoantigens/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Non-Fibrillar Collagens/genetics , Wnt Signaling Pathway , Collagen Type XVII
7.
Neurourol Urodyn ; 35(3): 377-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25557558

ABSTRACT

AIMS: We investigated the relationship between IL-1ß and morphological and functional changes following partial bladder outlet obstruction (pBOO). METHODS: Female wild-type C57/BL6 mice (WT) and IL-1ß-/- mice (KO) were used. Animals were sacrificed either 1 or 3 weeks after pBOO or sham surgery, and their bladders were harvested to determine bladder weight, for RT-PCR to measure interleukin-1ß (IL-1ß), insulin growth factor-1 (IGF-1), and transforming growth factor-ß (TGF-ß) levels, and for histological analysis with Hematoxylin-Eosin (HE) staining. Cystometry was performed on conscious animals 3 weeks after surgery to evaluate urodynamic parameters. IGF-1 was also administered intraperitoneally to KO with pBOO, and bladder weight was then investigated. RESULTS: IL-1ß-mRNA levels were significantly higher in WT-pBOO than in WT-sham. IGF-1-mRNA and TGF-ß-mRNA levels were also significantly higher in WT-pBOO than in WT-sham; however, these increases were smaller in KO-pBOO than in WT-pBOO. Bladder weight was significantly higher in WT-pBOO than in WT-sham, while increases in bladder weight were significantly suppressed in KO-pBOO. HE staining revealed the thickened bladder wall in WT-pBOO, and this phenomenon was less in KO-pBOO than in WT-pBOO. Regarding the urodynamic parameters examined, micturition pressure and bladder capacity were significantly higher in WT-pBOO than in WT-sham, but remained unchanged in KO-pBOO. The administration of IGF-1 to KO-pBOO led to similar increases in bladder weight and the thickened bladder wall as those observed in WT-pBOO. CONCLUSION: IL-1ß has the potential to induce bladder remodeling and deteriorate urodynamic parameters in pBOO.


Subject(s)
Cell Proliferation , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Urinary Bladder Neck Obstruction/metabolism , Urinary Bladder/metabolism , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Hypertrophy , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pressure , Signal Transduction , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Urinary Bladder/drug effects , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Urinary Bladder Neck Obstruction/genetics , Urinary Bladder Neck Obstruction/pathology , Urinary Bladder Neck Obstruction/physiopathology , Urination , Urodynamics
8.
Mol Cell Biol ; 35(11): 2007-23, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25825523

ABSTRACT

Wnt signaling pathways are tightly regulated by ubiquitination, and dysregulation of these pathways promotes tumorigenesis. It has been reported that the ubiquitin ligase RNF43 plays an important role in frizzled-dependent regulation of the Wnt/ß-catenin pathway. Here, we show that RNF43 suppresses both Wnt/ß-catenin signaling and noncanonical Wnt signaling by distinct mechanisms. The suppression of Wnt/ß-catenin signaling requires interaction between the extracellular protease-associated (PA) domain and the cysteine-rich domain (CRD) of frizzled and the intracellular RING finger domain of RNF43. In contrast, these N-terminal domains of RNF43 are not required for inhibition of noncanonical Wnt signaling, but interaction between the C-terminal cytoplasmic region of RNF43 and the PDZ domain of dishevelled is essential for this suppression. We further show the mechanism by which missense mutations in the extracellular portion of RNF43 identified in patients with tumors activate Wnt/ß-catenin signaling. Missense mutations of RNF43 change their localization from the endosome to the endoplasmic reticulum (ER), resulting in the failure of frizzled-dependent suppression of Wnt/ß-catenin signaling. However, these mutants retain the ability to suppress noncanonical Wnt signaling, probably due to interaction with dishevelled. RNF43 is also one of the potential target genes of Wnt/ß-catenin signaling. Our results reveal the molecular role of RNF43 and provide an insight into tumorigenesis.


Subject(s)
DNA-Binding Proteins/genetics , Oncogene Proteins/genetics , Signal Transduction/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Cell Line , Cell Line, Tumor , Cytoplasm/genetics , Cytoskeletal Proteins/genetics , Endoplasmic Reticulum/genetics , Endosomes/genetics , Frizzled Receptors/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mutation, Missense/genetics , RING Finger Domains/genetics , Trans-Activators/genetics , Ubiquitin-Protein Ligases , beta Catenin/genetics
9.
Nat Commun ; 6: 5941, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25575120

ABSTRACT

Regulation of transcription elongation by RNA polymerase II (Pol II) is a key regulatory step in gene transcription. Recently, the little elongation complex (LEC)-which contains the transcription elongation factor ELL/EAF-was found to be required for the transcription of Pol II-dependent small nuclear RNA (snRNA) genes. Here we show that the human Mediator subunit MED26 plays a role in the recruitment of LEC to a subset of snRNA genes through direct interaction of EAF and the N-terminal domain (NTD) of MED26. Loss of MED26 in cells decreases the occupancy of LEC at a subset of snRNA genes and results in a reduction in their transcription. Our results suggest that the MED26-NTD functions as a molecular switch in the exchange of TBP-associated factor 7 (TAF7) for LEC to facilitate the transition from initiation to elongation during transcription of a subset of snRNA genes.


Subject(s)
Mediator Complex/metabolism , Peptide Chain Elongation, Translational , RNA, Small Nuclear/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , DNA Polymerase II/metabolism , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Point Mutation , Protein Binding , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sf9 Cells , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism
10.
Neurosci Lett ; 512(1): 48-52, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22326927

ABSTRACT

Wnts are secreted glycoproteins that play important roles in embryonic development. Wnt2b is transiently expressed in the primitive streak (PS) during gastrulation and in several organs during organogenesis. To determine the biological function of Wnt2b during mouse development, we established a conditional null allele of Wnt2b. Mice lacking Wnt2b were viable, fertile, and displayed a normal life span, however, the olfactory bulb in adult Wnt2b mutant mice was significantly reduced in length. Our results suggest that Wnt2b primarily plays a supportive role in gastrulation and organogenesis, functioning redundantly with canonical Wnts, such as Wnt2, in numerous tissues.


Subject(s)
Olfactory Bulb/growth & development , Wnt Proteins/physiology , Animals , Brain/anatomy & histology , Brain/growth & development , Mice , Mice, Knockout , Olfactory Bulb/anatomy & histology , Phenotype , Wnt Proteins/genetics
11.
Mol Med ; 18: 587-97, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22331027

ABSTRACT

The nuclear factor (NF)-κB family of transcription factors regulates diverse cellular functions, including inflammation, oncogenesis and apoptosis. It was reported that A20 plays a critical role in the termination of NF-κB signaling after activation. Previously, we showed that Ymer interacts and collaborates with A20, followed by degradation of receptor-interacting protein (RIP) and attenuation of NF-κB signaling. Here we show the function of Ymer in regulation of several signaling pathways including NF-κB on the basis of results obtained by using Ymer transgenic (Ymer Tg) mice. Ymer Tg mice exhibited impaired immune responses, including NF-κB and mitogen-activated protein kinase (MAPK) activation, cell proliferation and cytokine production, to tumor necrosis factor (TNF)-α, polyI:C or lipopolysaccharide (LPS) stimulation. Ymer Tg mice were more resistant to LPS-induced septic shock than wild-type mice. Transgene of Ymer inhibited the onset of glomerulonephritis in lpr/lpr mice as an autoimmune disease model. In contrast to the inflammatory immune response to LPS, Fas-mediated cell death was strongly induced in liver cells of Ymer Tg mice in which Ymer is abundantly expressed. These findings suggest that Ymer acts as a regulator downstream of several receptors and that Ymer functions as a positive or negative regulator in a signaling pathway-dependent manner.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction , fas Receptor/metabolism , Animals , Apoptosis/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/immunology , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Thymocytes/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Biochim Biophys Acta ; 1813(6): 1245-53, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21463657

ABSTRACT

Ataxia-telangiectasia (AT) is an autosomal recessive genetic disease characterized by immunological deficiencies, neurological degeneration, developmental abnormalities and an increased risk of cancer. Ataxia-telangiectasia group D (ATDC) was initially described as a gene related to AT. Ataxia-telangiectasia group D, also known as TRIM29, is structurally a member of the tripartite motif (TRIM) family of proteins, some of which have been reported to be highly expressed in some human carcinomas, but the involvement of TRIM29 in carcinogenesis has not been fully elucidated. In this study, we found by using yeast two-hybrid screening that TRIM29 binds to Tip60, which has been reported as a cellular acetyltransferase protein. Overexpression of TRIM29 promoted degradation and changed localization of Tip60 and reduced acetylation of p53 at lysine 120 by Tip60, resulting in enhancement of cell growth and transforming activity. In addition, we found that TRIM29 suppresses apoptosis induced by UV irradiation in HCT116 cell lines. These findings suggest that TRIM29 functions as an oncogene that promotes tumor growth.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/genetics , Humans , Immunoblotting , Immunoprecipitation , Lysine/metabolism , Lysine Acetyltransferase 5 , Mice , NIH 3T3 Cells , Protein Binding , RNA Interference , Transcription Factors/genetics , Two-Hybrid System Techniques
13.
Kidney Int ; 79(9): 957-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21248711

ABSTRACT

Recent studies have identified several genes whose defects cause hereditary renal cystic diseases with most of the gene products located in the primary cilia. It has been suggested that primary cilia are involved in signaling pathways, defects of which result in abnormal cell proliferation and randomization of oriented cell division in the kidney leading to cyst formation. Mice with a mutation in the inv gene are a model for human nephronophthisis type 2 and develop multiple renal cysts. Inv protein (also called inversin) is located in the base of primary cilia and acts as a switch from canonical to non-canonical Wnt signaling. Here, we studied the orientation of cell division and proliferation in the kidneys of inv mutant mice, as its loss is thought to maintain activation of the canonical Wnt signaling. To establish if canonical signaling was involved in this process, we mated inv mutant with BATlacZ mice to measure canonical Wnt activity. Based on these reporter mice, nuclear localization and phosphorylation of ß-catenin, and responsiveness to Wnt ligands in inv mutant cells, we found that random oriented cell division is an initial event for renal tubule expansion and precedes cell proliferation. Thus, our results do not support the hypothesis that canonical Wnt signaling causes renal cyst development in these mice.


Subject(s)
Kidney Diseases, Cystic/etiology , Signal Transduction/physiology , Transcription Factors/physiology , Wnt Proteins/physiology , Animals , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Kidney/pathology , Mice , Mutation , Phosphorylation , Spindle Apparatus/physiology , beta Catenin/physiology
14.
Biochem Biophys Res Commun ; 404(1): 143-7, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21108931

ABSTRACT

The ubiquitin-proteasomal system plays a crucial role in oncogenesis in colorectal tissues. Recent studies have shown that stability of ß-catenin, which functions as an oncogene for colorectal cancer, is regulated by ubiquitin-mediated degradation. It has been reported that a putative E3 ubiquitin ligase, RNF43, is highly expressed in human colorectal carcinoma and that RNF43 promotes cell growth. However, the involvement of RNF43 in carcinogenesis has not been fully elucidated. In this study, we found by using yeast two-hybrid screening that RNF43 binds to NEDD-4-like ubiquitin-protein ligase-1 (NEDL1), which enhances pro-apoptotic activity by p53. In addition, we found that RNF43 also interacts with p53 and that RNF43 suppresses transcriptional activity of p53 in H1299 cells and attenuates apoptosis induced by ultraviolet irradiation. These findings suggest that RNF43 is associated with p53-mediated apoptosis in collaboration with NEDL1 in colorectal carcinogenesis.


Subject(s)
Carcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Nerve Tissue Proteins/genetics , Oncogene Proteins/genetics , Transcription, Genetic , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/genetics , Ultraviolet Rays
15.
Biochim Biophys Acta ; 1793(12): 1828-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909775

ABSTRACT

The androgen receptor (AR) is a ligand-dependent transcription factor that belongs to the family of nuclear receptors, and its activity is regulated by numerous AR coregulators. AR plays an important role in prostate development and cancer. In this study, we found that TRIM24/transcriptional intermediary factor 1alpha (TIF1alpha), which is known as a ligand-dependent nuclear receptor co-regulator, interacts with AR and enhances transcriptional activity of AR by dihydrotestosterone in prostate cancer cells. We showed that TRIM24 functionally interacts with TIP60, which acts as a coactivator of AR and synergizes with TIP60 in the transactivation of AR. We also showed that TRIM24 binds to bromodomain containing 7 (BRD7), which can negatively regulate cell proliferation and growth. A luciferase assay indicated that BRD7 represses the AR transactivation activity upregulated by TRIM24. These findings indicate that TRIM24 regulates AR-mediated transcription in collaboration with TIP60 and BRD7.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Androgens/pharmacology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/genetics , Dihydrotestosterone/pharmacology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Ligands , Lysine Acetyltransferase 5 , Male , Mice , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Repressor Proteins/genetics , Trans-Activators , Transcription Factors/genetics
16.
Mol Cancer Res ; 7(9): 1553-62, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723876

ABSTRACT

The ubiquitin-proteasome system has a crucial role in maintaining and regulating cellular homeostasis including carcinogenesis. UBE2Q2, also designated Ubci, is one of the ubiquitin-conjugating enzymes (E2), and it has been reported that mRNA of UBE2Q2 is highly expressed in human head and neck squamous cell carcinoma, particularly hypopharyngeal carcinoma. However, the involvement of UBE2Q2 in carcinogenesis has not been fully elucidated. Most cases of head and neck carcinoma are treated with cis-diamminedichloroplatinum (II; CDDP) or docetaxel, which are the most effective chemotherapeutic agents against squamous cell carcinomas. Nevertheless, some head and neck cancers develop resistance to these drugs, although the causes and mechanisms remain unknown. In this study, we found high expression levels of UBE2Q2 in human head and neck carcinoma cell lines and cancer tissues by using an anti-UBE2Q2 antibody at the protein level. We also found that the expression level of UBE2Q2 is decreased in cell lines and cancer tissues that have resistance to CDDP or docetaxel and in cancer tissues treated with CDDP or docetaxel. Furthermore, we found that overexpression of UBE2Q2 affects cell proliferation and anchorage-independent cell growth. These findings suggest that UBE2Q2 is a novel oncosuppressor that inhibits tumor growth and is related to the resistance to anticarcinoma agents and that UBE2Q2 likely functions as a novel diagnostic tool and a potentially therapeutic target for head and neck squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cytoskeletal Proteins/biosynthesis , Head and Neck Neoplasms/enzymology , Ubiquitin-Conjugating Enzymes/biosynthesis , Animals , Antineoplastic Agents/pharmacology , COS Cells , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , Chlorocebus aethiops , Cisplatin/pharmacology , Cytoskeletal Proteins/genetics , Docetaxel , Down-Regulation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , HeLa Cells , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Mice , NIH 3T3 Cells , Signal Transduction , Taxoids/pharmacology , Ubiquitin-Conjugating Enzymes/genetics , src-Family Kinases/metabolism
17.
Biochem Biophys Res Commun ; 388(2): 422-7, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19665990

ABSTRACT

Tripartite motif-containing protein (TRIM) family proteins are involved in a broad range of biological processes and, consistently, their alterations result in diverse pathological conditions such as genetic diseases, viral infection and cancer development. In this study, we found that one of the TRIM family proteins, TRIM31, is highly expressed in the gastrointestinal tract and interacts with p52(Shc), one of the signal transducers. We also found by a binding assay that almost the whole region other than the RING domain is required for the binding to p52(Shc) but found by pulse-chase analysis that overexpression of TRIM31 does not affect the stability of p52(Shc). Moreover, we found that overexpression of TRIM31 suppresses anchorage-independent cell growth induced by the active form of c-Src. These results suggest that TRIM31 attenuates c-Src signaling via p52(Shc) under anchorage-independent growth conditions and is potentially associated with growth activity of cells in the gastrointestinal tract.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Gastrointestinal Tract/metabolism , Nuclear Proteins/metabolism , Shc Signaling Adaptor Proteins/metabolism , src-Family Kinases/metabolism , Animals , Carrier Proteins/genetics , Gastrointestinal Tract/cytology , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/genetics , Protein Stability , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tripartite Motif Proteins , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases , src-Family Kinases/genetics
18.
Dev Cell ; 17(2): 290-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19686689

ABSTRACT

Patterning of the primitive foregut promotes appropriate organ specification along its anterior-posterior axis. However, the molecular pathways specifying foregut endoderm progenitors are poorly understood. We show here that Wnt2/2b signaling is required to specify lung endoderm progenitors within the anterior foregut. Embryos lacking Wnt2/2b expression exhibit complete lung agenesis and do not express Nkx2.1, the earliest marker of the lung endoderm. In contrast, other foregut endoderm-derived organs, including the thyroid, liver, and pancreas, are correctly specified. The phenotype observed is recapitulated by an endoderm-restricted deletion of beta-catenin, demonstrating that Wnt2/2b signaling through the canonical Wnt pathway is required to specify lung endoderm progenitors within the foregut. Moreover, activation of canonical Wnt/beta-catenin signaling results in the reprogramming of esophagus and stomach endoderm to a lung endoderm progenitor fate. Together, these data reveal that canonical Wnt2/2b signaling is required for the specification of lung endoderm progenitors in the developing foregut.


Subject(s)
Digestive System , Lung , Signal Transduction/physiology , Stem Cells/physiology , Wnt Proteins/metabolism , Wnt2 Protein/metabolism , beta Catenin/metabolism , Animals , Body Patterning/physiology , Digestive System/anatomy & histology , Digestive System/embryology , Endoderm/cytology , Endoderm/physiology , Lung/anatomy & histology , Lung/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cells/cytology , Wnt Proteins/genetics , Wnt2 Protein/genetics , beta Catenin/genetics
19.
Development ; 136(3): 367-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19141668

ABSTRACT

beta-catenin signaling is one of the key factors regulating the fate of hair follicles (HFs). To elucidate the regulatory mechanism of embryonic HF fate determination during epidermal development/differentiation, we analyzed conditional mutant mice with keratinocytes expressing constitutively active beta-catenin (K5-Cre Catnb(ex3)fl/+). The mutant mice developed scaly skin with a thickened epidermis and showed impaired epidermal stratification. The hair shaft keratins were broadly expressed in the epidermis but there was no expression of the terminal differentiation markers K1 and loricrin. Hair placode markers (Bmp2 and Shh) and follicular dermal condensate markers (noggin, patched 1 and Pdgfra) were expressed throughout the epidermis and the upper dermis, respectively. These results indicate that the embryonic epidermal keratinocytes have switched extensively to the HF fate. A series of genetic studies demonstrated that the epidermal switching to HF fate was suppressed by introducing the conditional mutation K5-Cre Catnb(ex3)fl/+Shhfl/- (with additional mutation of Shh signaling) or K5-Cre Catnb(ex3)fl/+BmprIAfl/fl (with additional mutation of Bmp signaling). These results demonstrate that Wnt/beta-catenin signaling relayed through Shh and Bmp signals is the principal regulatory mechanism underlying the HF cell fate change. Assessment of Bmp2 promoter activities suggested a putative regulation by beta-catenin signaling relayed by Shh signaling towards Bmp2. We also found that Shh protein expression was increased and expanded in the epidermis of K5-Cre Catnb(ex3)fl/+BmprIAfl/fl mice. These results indicate the presence of growth factor signal cross-talk involving beta-catenin signaling, which regulates the HF fate.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Epidermal Cells , Hair Follicle/cytology , Hedgehog Proteins/physiology , beta Catenin/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Cell Differentiation/physiology , Epidermis/embryology , Gene Expression Regulation, Developmental , Hair Follicle/embryology , Hair Follicle/physiology , Hedgehog Proteins/genetics , Keratinocytes/cytology , Keratinocytes/physiology , Keratins, Hair-Specific/metabolism , Mice , Mice, Mutant Strains , Mutation , Signal Transduction/physiology , beta Catenin/genetics
20.
Biochem Biophys Res Commun ; 378(4): 744-9, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19059208

ABSTRACT

Cytoplasmic zinc finger protein A20 functionally dampens inflammatory signals and apoptosis via inhibition of NF-kappaB activation. We have reported that Ymer interacts with A20 and lysine (K)-63-linked polyubiquitin chain and that Ymer inhibits NF-kappaB signaling in collaboration with A20. It has also been reported that Ymer is phosphorylated by EGF stimulation. We found that Ymer was considerably phosphorylated on tyrosine residues also via Src family kinases such as Lck. A luciferase reporter assay showed that mutation of tyrosines on Ymer (YmerY217/279/304F) results in loss of the inhibitory activity for NF-kappaB signaling. Furthermore, a soft agar colony formation assay showed that the combination of SrcY527F and YmerY217/279/304F has no ability for anchorage-independent growth, suggesting that tyrosine phosphorylation of Ymer is important for inhibition of the NF-kappaB-mediated apoptotic pathway. These findings demonstrate that Ymer is likely to be a negative regulator for the NF-kappaB signaling pathway.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Tyrosine/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Phosphorylation , Signal Transduction , Tyrosine/genetics , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...