Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 17586, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266461

ABSTRACT

The transcription factor hepatocyte nuclear factor 1ß (HNF-1ß) is essential for normal development of the kidney and other epithelial organs. In the developing mouse kidney, HNF-1ß is required for the differentiation and patterning of immature nephrons and branching morphogenesis of the ureteric bud (UB). Here, we used ChIP-sequencing (ChIP-seq) and RNA sequencing (RNA-seq) to identify genes that are regulated by HNF-1ß in embryonic mouse kidneys. ChIP-seq revealed that HNF-1ß binds to 8284 sites in chromatin from E14.5 mouse kidneys. Comparison with previous ATAC-seq and histone modification studies showed that HNF-1ß binding peaks colocalized with open chromatin and epigenetic marks of transcriptional activation (H3K27 acetylation, H3K4 trimethylation, H3K4 monomethylation), indicating that the binding sites were functional. To investigate the relationship between HNF-1ß binding and HNF-1ß-dependent gene regulation, RNA-seq was performed on UB cells purified from wild-type and HNF-1ß mutant embryonic kidneys. A total of 1632 genes showed reduced expression in HNF-1ß-deficient UB cells, and 485 genes contained nearby HNF-1ß binding sites indicating that they were directly activated by HNF-1ß. Conversely, HNF-1ß directly repressed the expression of 526 genes in the UB. Comparison with snATAC-seq analysis of UB-derived cells showed that both HNF-1ß-dependent activation and repression correlated with chromatin accessibility. Pathway analysis revealed that HNF-1ß binds near 68 axon guidance genes in the developing kidney. RNA-seq analysis showed that Nrp1, Sema3c, Sema3d, Sema6a, and Slit2 were activated by HNF-1ß, whereas Efna1, Epha3, Epha4, Epha7, Ntn4, Plxna2, Sema3a, Sema4b, Slit3, Srgap1, Unc5c and Unc5d were repressed by HNF-1ß. RNAscope in situ hybridization showed that Nrp1, Sema3c, Sema3d, Sema6a, and Slit2 were expressed in wild-type UB and were dysregulated in HNF-1ß mutant UB. These studies show that HNF-1ß directly regulates the expression of multiple axon guidance genes in the developing mouse kidney. Dysregulation of axon guidance genes may underlie kidney defects in HNF-1ß mutant mice.


Subject(s)
Axon Guidance , Hepatocyte Nuclear Factor 1-beta , Animals , Mice , Axon Guidance/genetics , Chromatin/genetics , Chromatin/metabolism , Ephrin-A1/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Semaphorin-3A/genetics , Semaphorins/genetics , Transcription Factors/metabolism
2.
Cell Signal ; 71: 109568, 2020 07.
Article in English | MEDLINE | ID: mdl-32068086

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is a DNA-binding transcription factor that is essential for normal kidney development. Mutations of HNF1B in humans produce cystic kidney diseases, including renal cysts and diabetes, multicystic dysplastic kidneys, glomerulocystic kidney disease, and autosomal dominant tubulointerstitial kidney disease. Expression of HNF1B is reduced in cystic kidneys from humans with ADPKD, and HNF1B has been identified as a modifier gene in PKD. Genome-wide analysis of chromatin binding has revealed that HNF-1ß directly regulates the expression of known PKD genes, such as PKHD1 and PKD2, as well as genes involved in PKD pathogenesis, including cAMP-dependent signaling, renal fibrosis, and Wnt signaling. In addition, a role of HNF-1ß in regulating the expression of noncoding RNAs (microRNAs and long noncoding RNAs) has been identified. These findings indicate that HNF-1ß regulates a transcriptional and post-transcriptional network that plays a central role in renal cystogenesis.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Polycystic Kidney Diseases/metabolism , Animals , Hepatocyte Nuclear Factor 1-beta/chemistry , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Models, Biological , Mutation/genetics , Polycystic Kidney Diseases/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Signal Transduction
3.
J Biol Chem ; 295(51): 17560-17572, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33453998

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is a tissue-specific transcription factor that is required for normal kidney development and renal epithelial differentiation. Mutations of HNF-1ß produce congenital kidney abnormalities and inherited renal tubulopathies. Here, we show that ablation of HNF-1ß in mIMCD3 renal epithelial cells results in activation of ß-catenin and increased expression of lymphoid enhancer-binding factor 1 (LEF1), a downstream effector in the canonical Wnt signaling pathway. Increased expression and nuclear localization of LEF1 are also observed in cystic kidneys from Hnf1b mutant mice. Expression of dominant-negative mutant HNF-1ß in mIMCD3 cells produces hyperresponsiveness to exogenous Wnt ligands, which is inhibited by siRNA-mediated knockdown of Lef1. WT HNF-1ß binds to two evolutionarily conserved sites located 94 and 30 kb from the mouse Lef1 promoter. Ablation of HNF-1ß decreases H3K27 trimethylation repressive marks and increases ß-catenin occupancy at a site 4 kb upstream to Lef1. Mechanistically, WT HNF-1ß recruits the polycomb-repressive complex 2 that catalyzes H3K27 trimethylation. Deletion of the ß-catenin-binding domain of LEF1 in HNF-1ß-deficient cells abolishes the increase in Lef1 transcription and decreases the expression of downstream Wnt target genes. The canonical Wnt target gene, Axin2, is also a direct transcriptional target of HNF-1ß through binding to negative regulatory elements in the gene promoter. These findings demonstrate that HNF-1ß regulates canonical Wnt target genes through long-range effects on histone methylation at Wnt enhancers and reveal a new mode of active transcriptional repression by HNF-1ß.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Wnt Signaling Pathway , Animals , Axin Protein/genetics , Axin Protein/metabolism , Binding Sites , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/genetics , Histones/metabolism , Kidney/cytology , Lymphoid Enhancer-Binding Factor 1/antagonists & inhibitors , Lymphoid Enhancer-Binding Factor 1/genetics , Methylation , Mice , Mice, Knockout , Mutagenesis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Regulatory Elements, Transcriptional/genetics , Wnt3A Protein/metabolism , beta Catenin/metabolism
4.
Proc Natl Acad Sci U S A ; 116(48): 24133-24142, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31712448

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is a tissue-specific transcription factor that is essential for normal kidney development and renal tubular function. Mutations of HNF-1ß produce cystic kidney disease, a phenotype associated with deregulation of canonical (ß-catenin-dependent) Wnt signaling. Here, we show that ablation of HNF-1ß in mIMCD3 renal epithelial cells produces hyperresponsiveness to Wnt ligands and increases expression of Wnt target genes, including Axin2, Ccdc80, and Rnf43 Levels of ß-catenin and expression of Wnt target genes are also increased in HNF-1ß mutant mouse kidneys. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) in wild-type and mutant cells showed that ablation of HNF-1ß increases by 6-fold the number of sites on chromatin that are occupied by ß-catenin. Remarkably, 50% of the sites that are occupied by ß-catenin in HNF-1ß mutant cells colocalize with HNF-1ß-occupied sites in wild-type cells, indicating widespread reciprocal binding. We found that the Wnt target genes Ccdc80 and Rnf43 contain a composite DNA element comprising a ß-catenin/lymphoid enhancer binding factor (LEF) site overlapping with an HNF-1ß half-site. HNF-1ß and ß-catenin/LEF compete for binding to this element, and thereby HNF-1ß inhibits ß-catenin-dependent transcription. Collectively, these studies reveal a mechanism whereby a transcription factor constrains canonical Wnt signaling through direct inhibition of ß-catenin/LEF chromatin binding.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Cell Line , Enhancer Elements, Genetic , Epithelial Cells/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Medulla/cytology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice, Knockout , Mice, Transgenic , Mutation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , beta Catenin/metabolism
5.
J Am Soc Nephrol ; 29(10): 2493-2509, 2018 10.
Article in English | MEDLINE | ID: mdl-30097458

ABSTRACT

BACKGROUND: Mutation of HNF1B, the gene encoding transcription factor HNF-1ß, is one cause of autosomal dominant tubulointerstitial kidney disease, a syndrome characterized by tubular cysts, renal fibrosis, and progressive decline in renal function. HNF-1ß has also been implicated in epithelial-mesenchymal transition (EMT) pathways, and sustained EMT is associated with tissue fibrosis. The mechanism whereby mutated HNF1B leads to tubulointerstitial fibrosis is not known. METHODS: To explore the mechanism of fibrosis, we created HNF-1ß-deficient mIMCD3 renal epithelial cells, used RNA-sequencing analysis to reveal differentially expressed genes in wild-type and HNF-1ß-deficient mIMCD3 cells, and performed cell lineage analysis in HNF-1ß mutant mice. RESULTS: The HNF-1ß-deficient cells exhibited properties characteristic of mesenchymal cells such as fibroblasts, including spindle-shaped morphology, loss of contact inhibition, and increased cell migration. These cells also showed upregulation of fibrosis and EMT pathways, including upregulation of Twist2, Snail1, Snail2, and Zeb2, which are key EMT transcription factors. Mechanistically, HNF-1ß directly represses Twist2, and ablation of Twist2 partially rescued the fibroblastic phenotype of HNF-1ß mutant cells. Kidneys from HNF-1ß mutant mice showed increased expression of Twist2 and its downstream target Snai2. Cell lineage analysis indicated that HNF-1ß mutant epithelial cells do not transdifferentiate into kidney myofibroblasts. Rather, HNF-1ß mutant epithelial cells secrete high levels of TGF-ß ligands that activate downstream Smad transcription factors in renal interstitial cells. CONCLUSIONS: Ablation of HNF-1ß in renal epithelial cells leads to the activation of a Twist2-dependent transcriptional network that induces EMT and aberrant TGF-ß signaling, resulting in renal fibrosis through a cell-nonautonomous mechanism.


Subject(s)
Gout/genetics , Gout/pathology , Hepatocyte Nuclear Factor 1-beta/genetics , Hyperuricemia/genetics , Hyperuricemia/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Animals , Cell Line , Cell Lineage/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Fibrosis , Genes, Dominant , Gout/metabolism , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Hyperuricemia/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Male , Mice , Mice, Transgenic , Mutation , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/deficiency , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
6.
J Biol Chem ; 293(24): 9388-9398, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29716997

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a debilitating disease that is characterized by the accumulation of numerous fluid-filled cysts in the kidney. ADPKD is primarily caused by mutations in two genes, PKD1 and PKD2 Long noncoding RNAs (lncRNA), defined by a length >200 nucleotides and absence of a long ORF, have recently emerged as epigenetic regulators of development and disease; however, their involvement in PKD has not been explored previously. Here, we performed deep RNA-Seq to identify lncRNAs that are dysregulated in two orthologous mouse models of ADPKD (kidney-specific Pkd1 and Pkd2 mutant mice). We identified a kidney-specific, evolutionarily conserved lncRNA called Hoxb3os that was down-regulated in cystic kidneys from Pkd1 and Pkd2 mutant mice. The human ortholog HOXB3-AS1 was down-regulated in cystic kidneys from ADPKD patients. Hoxb3os was highly expressed in renal tubules in adult WT mice, whereas its expression was lost in the cyst epithelium of mutant mice. To investigate the function of Hoxb3os, we utilized CRISPR/Cas9 to knock out its expression in mIMCD3 cells. Deletion of Hoxb3os resulted in increased phosphorylation of mTOR and its downstream targets, including p70 S6 kinase, ribosomal protein S6, and the translation repressor 4E-BP1. Consistent with activation of mTORC1 signaling, Hoxb3os mutant cells displayed increased mitochondrial respiration. The Hoxb3os mutant phenotype was partially rescued upon re-expression of Hoxb3os in knockout cells. These findings identify Hoxb3os as a novel lncRNA that is down-regulated in ADPKD and regulates mTOR signaling and mitochondrial respiration.


Subject(s)
Polycystic Kidney, Autosomal Dominant/genetics , RNA, Long Noncoding/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Mice, Inbred C57BL , Mutation , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/genetics
7.
Nucleic Acids Res ; 43(12): 5880-97, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25908785

ABSTRACT

Androgen receptor (AR) variants (AR-Vs) expressed in prostate cancer (PCa) lack the AR ligand binding domain (LBD) and function as constitutively active transcription factors. AR-V expression in patient tissues or circulating tumor cells is associated with resistance to AR-targeting endocrine therapies and poor outcomes. Here, we investigated the mechanisms governing chromatin binding of AR-Vs with the goal of identifying therapeutic vulnerabilities. By chromatin immunoprecipitation and sequencing (ChIP-seq) and complementary biochemical experiments, we show that AR-Vs display a binding preference for the same canonical high-affinity androgen response elements (AREs) that are preferentially engaged by AR, albeit with lower affinity. Dimerization was an absolute requirement for constitutive AR-V DNA binding and transcriptional activation. Treatment with the bromodomain and extraterminal (BET) inhibitor JQ1 resulted in inhibition of AR-V chromatin binding and impaired AR-V driven PCa cell growth in vitro and in vivo. Importantly, this was associated with a novel JQ1 action of down-regulating AR-V transcript and protein expression. Overall, this study demonstrates that AR-Vs broadly restore AR chromatin binding events that are otherwise suppressed during endocrine therapy, and provides pre-clinical rationale for BET inhibition as a strategy for inhibiting expression and chromatin binding of AR-Vs in PCa.


Subject(s)
Chromatin/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects , Animals , Antineoplastic Agents/therapeutic use , Azepines/pharmacology , Benzamides , Cell Line , Cell Line, Tumor , Dimerization , Male , Mice, Nude , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Response Elements , Triazoles/pharmacology
8.
Adv Pharmacol ; 70: 327-66, 2014.
Article in English | MEDLINE | ID: mdl-24931201

ABSTRACT

Prostate cancer (PCa) is the most frequently diagnosed cancer in the United States. The androgen receptor (AR) signaling axis is central to all stages of PCa pathophysiology and serves as the main target for endocrine-based therapy. The most advanced stage of the disease, castration-resistant prostate cancer (CRPC), is presently incurable and accounts for most PCa mortality. In this chapter, we highlight the mechanisms by which the AR signaling axis can bypass endocrine-targeted therapies and drive progression of CRPC. These mechanisms include alterations in growth factor, cytokine, and inflammatory signaling pathways, altered expression or activity of transcriptional coregulators, AR point mutations, and AR gene amplification leading to AR protein overexpression. Additionally, we will discuss the mechanisms underlying the synthesis of constitutively active AR splice variants (AR-Vs) lacking the COOH-terminal ligand-binding domain, as well as the role and regulation of AR-Vs in supporting therapeutic resistance in CRPC. Finally, we summarize the ongoing development of inhibitors targeting discrete AR functional domains as well as the status of new biomarkers for monitoring the AR signaling axis in patients.


Subject(s)
Mutation/genetics , Receptors, Androgen/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Gene Amplification , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/genetics , Receptors, Androgen/chemistry , Signal Transduction
9.
Cancer Res ; 73(2): 483-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23117885

ABSTRACT

Persistent androgen receptor (AR) transcriptional activity underlies resistance to AR-targeted therapy and progression to lethal castration-resistant prostate cancer (CRPC). Recent success in retargeting persistent AR activity with next generation androgen/AR axis inhibitors such as enzalutamide (MDV3100) has validated AR as a master regulator during all stages of disease progression. However, resistance to next generation AR inhibitors limits therapeutic efficacy for many patients. One emerging mechanism of CRPC progression is AR gene rearrangement, promoting synthesis of constitutively active truncated AR splice variants (AR-V) that lack the AR ligand-binding domain. In this study, we show that cells with AR gene rearrangements expressing both full-length and AR-Vs are androgen independent and enzalutamide resistant. However, selective knock-down of AR-V expression inhibited androgen-independent growth and restored responsiveness to androgens and antiandrogens. In heterogeneous cell populations, AR gene rearrangements marked individual AR-V-dependent cells that were resistant to enzalutamide. Gene expression profiling following knock-down of full-length AR or AR-Vs showed that AR-Vs drive resistance to AR-targeted therapy by functioning as constitutive and independent effectors of the androgen/AR transcriptional program. Further, mitotic genes deemed previously to be unique AR-V targets were found to be biphasic targets associated with a proliferative level of signaling output from either AR-Vs or androgen-stimulated AR. Overall, these studies highlight AR-Vs as key mediators of persistent AR signaling and resistance to the current arsenal of conventional and next generation AR-directed therapies, advancing the concept of AR-Vs as therapeutic targets in advanced disease.


Subject(s)
Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Isoforms , Receptors, Androgen/genetics , Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Benzamides , Castration , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Rearrangement , Humans , Male , Neoplasms, Hormone-Dependent/genetics , Nitriles , Phenylthiohydantoin/therapeutic use
10.
J Biol Chem ; 287(23): 19736-49, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22532567

ABSTRACT

Synthesis of truncated androgen receptor (AR) splice variants has emerged as an important mechanism of prostate cancer (PCa) resistance to AR-targeted therapy and progression to a lethal castration-resistant phenotype. However, the precise role of these factors at this stage of the disease is not clear due to loss of multiple COOH-terminal AR protein domains, including the canonical nuclear localization signal (NLS) in the AR hinge region. Despite loss of this NLS, we show that diverse truncated AR variant species have a basal level of nuclear localization sufficient for ligand-independent transcriptional activity. Whereas full-length AR requires Hsp90 and importin-ß for active nuclear translocation, basal nuclear localization of truncated AR variants is independent of these classical signals. For a subset of truncated AR variants, this basal level of nuclear import can be augmented by unique COOH-terminal sequences that reconstitute classical AR NLS activity. However, this property is separable from ligand-independent transcriptional activity. Therefore, the AR splice variant core consisting of the AR NH(2)-terminal domain and DNA binding domain is sufficient for nuclear localization and androgen-independent transcriptional activation of endogenous AR target genes. Indeed, we show that truncated AR variants with nuclear as well as nuclear/cytoplasmic localization patterns can drive androgen-independent growth of PCa cells. Together, our data demonstrate that diverse truncated AR species with varying efficiencies of nuclear localization can contribute to castration-resistant PCa pathology by driving persistent ligand-independent AR transcriptional activity.


Subject(s)
Alternative Splicing , Cell Nucleus/metabolism , Neoplasm Proteins/metabolism , Nuclear Localization Signals , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Active Transport, Cell Nucleus/genetics , Animals , COS Cells , Cell Nucleus/genetics , Cell Nucleus/pathology , Chlorocebus aethiops , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Transcription, Genetic/genetics , beta Karyopherins/genetics , beta Karyopherins/metabolism
11.
Biochem J ; 408(2): 259-66, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17711404

ABSTRACT

Cidea, one of three members of the CIDE (cell-death-inducing DNA-fragmentation-factor-45-like effector) family of proteins, is highly enriched in brown adipose tissue, in which it plays a critical role in adaptive thermogenesis and fat accumulation. Cidea-null mice have increased energy expenditure with resistance to high-fat-diet-induced obesity and diabetes. However, little is known as to how the Cidea protein is regulated. In the present study we show that Cidea is a short-lived protein as measured by cycloheximide-based protein chase experiments in different cell lines or in differentiated brown adipocytes. Proteasome inhibitors specifically increased the stability of both transfected and endogenous Cidea protein. Furthermore, Cidea protein was found to be polyubiquitinated when overexpressed in different culture cells as well as in differentiated mature brown adipocytes. Extensive mutational analysis of individual lysine residues revealed that ubiquitinated lysine residues are located in the N-terminal region of Cidea, as alteration of these lysine residues to alanine (N-5KA mutant) renders Cidea much more stable when compared with wild-type or C-terminal lysine-less mutant (C-5KA). Furthermore, K23 (Lys23) within the N-terminus of the Cidea was identified as the major contributor to its polyubiquitination signal and the protein instability. Taken together, the results of our study demonstrated that the ubiquitin-proteasome system confers an important post-translational modification that controls the protein stability of Cidea.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/genetics , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Mice , Molecular Sequence Data , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/physiology , Proteasome Inhibitors , Signal Transduction/genetics , Thermodynamics , Ubiquitin/genetics , Ubiquitin/physiology
12.
Dev Cell ; 13(2): 268-82, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17681137

ABSTRACT

Axin is a scaffold protein that controls multiple important pathways, including the canonical Wnt pathway and JNK signaling. Here we have identified an Axin-interacting protein, Aida, which blocks Axin-mediated JNK activation by disrupting Axin homodimerization. During investigation of in vivo functions of Axin/JNK signaling and aida in development, it was found that Axin, besides ventralizing activity by facilitating beta-catenin degradation, possesses a dorsalizing activity that is mediated by Axin-induced JNK activation. This dorsalizing activity is repressed when aida is overexpressed in zebrafish embryos. Whereas Aida-MO injection leads to dorsalized embryos, JNK-MO and MKK4-MO can ventralize embryos. The anti-dorsalization activity of aida is conferred by its ability to block Axin-mediated JNK activity. We further demonstrate that dorsoventral patterning regulated by Axin/JNK signaling is independent of maternal or zygotic Wnt signaling. We have thus identified a dorsalization pathway that is exerted by Axin/JNK signaling and its inhibitor Aida during vertebrate embryogenesis.


Subject(s)
Body Patterning , Carrier Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Repressor Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , beta Catenin/metabolism , Animals , Axin Protein , Body Patterning/drug effects , COS Cells , Cell Line , Chlorocebus aethiops , Dimerization , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/enzymology , Enzyme Activation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mice , Oligonucleotides, Antisense/pharmacology , Phenotype , Protein Binding/drug effects , Repressor Proteins/chemistry , Wnt Proteins/metabolism , Zebrafish/metabolism
13.
EMBO J ; 25(8): 1646-58, 2006 Apr 19.
Article in English | MEDLINE | ID: mdl-16601693

ABSTRACT

TGF-beta signaling involves a wide array of signaling molecules and multiple controlling events. Scaffold proteins create a functional proximity of signaling molecules and control the specificity of signal transduction. While many components involved in the TGF-beta pathway have been elucidated, little is known about how those components are coordinated by scaffold proteins. Here, we show that Axin activates TGF-beta signaling by forming a multimeric complex consisting of Smad7 and ubiquitin E3 ligase Arkadia. Axin depends on Arkadia to facilitate TGF-beta signaling, as their small interfering RNAs reciprocally abolished the stimulatory effect on TGF-beta signaling. Specific knockdown of Axin or Arkadia revealed that Axin and Arkadia cooperate with each other in promoting Smad7 ubiquitination. Pulse-chase experiments further illustrated that Axin significantly decreased the half-life of Smad7. Axin also induces nuclear export of Smad7. Interestingly, Axin associates with Arkadia and Smad7 independently of TGF-beta signal, in contrast to its transient association with inactive Smad3. However, coexpression of Wnt-1 reduced Smad7 ubiquitination by downregulating Axin levels, underscoring the importance of Axin as an intrinsic regulator in TGF-beta signaling.


Subject(s)
Repressor Proteins/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta/physiology , Ubiquitin-Protein Ligases/physiology , Ubiquitin/physiology , Active Transport, Cell Nucleus , Animals , Axin Protein , Cell Line , Cell Nucleus/metabolism , Humans , Mice , Mutation , Phosphorylation , Protein Binding , Repressor Proteins/genetics , Signal Transduction , Smad3 Protein/metabolism , Smad7 Protein/genetics , Ubiquitin-Protein Ligases/genetics , Wnt1 Protein/metabolism
14.
FEBS J ; 272(15): 3967-74, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16045767

ABSTRACT

Staphylococcal nuclease is a single domain protein with 149 amino acids. It has no disulfide bonds, which makes it a simple model for the study of protein folding. In this study, 20 mutants of this protein were generated each with a single base substitution of glycine for negatively charged glutamic acid or aspartic acid. Using differential scanning microcalorimetry in thermal denaturation experiments, we identified two mutants, E75G and E129G, having approximately 43% and 44%, respectively, lower DeltaH(cal) values than the wild-type protein. Furthermore, two mutants, E75Q and E129Q, were created and the results imply that substitution of the Gly residue has little influence on destabilization of the secondary structure that leads to the large perturbation of the tertiary protein structure stability. Two local stable areas formed by the charge-charge interactions around E75 and E129 with particular positively charged amino acids are thus identified as being significant in maintenance of the three-dimensional structure of the protein.


Subject(s)
Aspartic Acid/metabolism , Glutamic Acid/metabolism , Micrococcal Nuclease/metabolism , Protein Folding , Aspartic Acid/chemistry , Aspartic Acid/genetics , Calorimetry, Differential Scanning , Circular Dichroism , Enzyme Stability/physiology , Glutamic Acid/chemistry , Glutamic Acid/genetics , Micrococcal Nuclease/chemistry , Micrococcal Nuclease/genetics , Mutation , Protein Denaturation/physiology , Protein Structure, Tertiary , Spectrometry, Fluorescence , Temperature , Thermodynamics
15.
J Cell Sci ; 117(Pt 20): 4673-80, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15371523

ABSTRACT

Tumor necrosis factor (TNF)-alpha induces caspase-independent cell death in the fibrosarcoma cell line L929. This cell death has a necrotic phenotype and is dependent on production of reactive oxygen species (ROS) in the mitochondria. To identify genes involved in this TNF-induced, ROS-dependent cell death pathway, we utilized retrovirus insertion-mediated random mutagenesis to generate TNF-resistant L929 cell lines and we subsequently identified genes whose mutations are responsible for the TNF-resistant phenotype. In one such resistant line, beta-actin was disrupted by viral insertion, and subsequent reconstitution of beta-actin expression levels in the mutant line Actin(mut) restored its sensitivity to TNF. Resistance to TNF in Actin(mut) cells is signal specific since the sensitivity to other death stimuli is either unchanged or even increased. Comparable NF-kappaB activation and p38 phosphorylation in TNF-treated wild-type and Actin(mut) cells also indicates that reduced expression of actin only selectively blocked some of the TNF-induced cellular changes. Actin cleavage involved in apoptosis does not occur in TNF-treated L929 cell death, as in HeLa cells. Consistent over-expression of a caspase-cleaved product, a 15 kDa actin fragment, had no effect on TNF-induced necrosis of L929 cell. By contrast, TNF-induced mitochondria clustering and ROS production were dramatically reduced in Actin(mut) cells, indicating that actin-deficiency-mediated TNF resistance is most likely due to impaired mitochondrial responses to TNF stimulation. Our findings suggest that a full complement of actin is required for transduction of a cell death signal to mitochondria in TNF-treated L929 cells.


Subject(s)
Actins/metabolism , Caspases/metabolism , Cell Death/genetics , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Actins/genetics , Animals , Cell Death/physiology , Cell Line , Enzyme Activation , Humans , Mice , Mitochondria/metabolism , Mutation , NF-kappa B/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Biol Chem ; 277(20): 17706-12, 2002 May 17.
Article in English | MEDLINE | ID: mdl-11884395

ABSTRACT

Axin uses different combinations of functional domains in down-regulation of the Wnt pathway and activation of the MEKK1/JNK pathway. We are interested in the elucidation of the functional switch of Axin. In the present study, we show that the Wnt activator CKIepsilon, but not CKIIalpha, Frat1, LRP5, or LRP6, inhibited Axin-mediated JNK activation. We also found that both CKIalpha and CKIepsilon interacted with Axin, whereas CKIIalpha did not bind to Axin and had no effect on Axin-mediated JNK activity even though CKIIalpha has also been suggested to be an activator for the Wnt pathway. The COOH-terminal region and the MEKK1-interacting domain of Axin are important for CKIalpha-Axin and CKIepsilon-Axin interaction. We further demonstrated that CKIepsilon and CKIalpha binding to Axin excluded MEKK1 binding, indicating that a competitive physical occupancy may underlie the inhibitory effect. Moreover, our data indicated that CKIepsilon kinase activity plays an additive role in this effect. Taken together, we have demonstrated that CKI and CKII exhibit differential effects on Axin-MEKK1 interaction and Axin-mediated JNK activation. Furthermore, our data suggest that CKI may provide a possible switch mechanism for Axin function in the regulation of Wnt and JNK pathways.


Subject(s)
MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins , Zebrafish Proteins , Animals , Axin Protein , Casein Kinase II , Casein Kinases , Cells, Cultured , Enzyme Activation , Humans , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Mice , Peptide Mapping , Protein Binding , Protein Serine-Threonine Kinases/physiology , Wnt Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...