Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 295(14): 4617-4630, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32115405

ABSTRACT

Specificity protein 1 (Sp1) is an important transcription factor implicated in numerous cellular processes. However, whether Sp1 is involved in the regulation of RNA polymerase III (Pol III)-directed gene transcription in human cells remains unknown. Here, we first show that filamin A (FLNA) represses Sp1 expression as well as expression of TFIIB-related factor 1 (BRF1) and general transcription factor III C subunit 2 (GTF3C2) in HeLa, 293T, and SaOS2 cell lines stably expressing FLNA-silencing shRNAs. Both BRF1 promoter 4 (BRF1P4) and GTF3C2 promoter 2 (GTF3C2P2) contain putative Sp1-binding sites, suggesting that Sp1 affects Pol III gene transcription by regulating BRF1 and GTF3C2 expression. We demonstrate that Sp1 knockdown inhibits Pol III gene transcription, BRF1 and GTF3C2 expression, and the proliferation of 293T and HeLa cells, whereas Sp1 overexpression enhances these activities. We obtained a comparable result in a cell line in which both FLNA and Sp1 were depleted. These results indicate that Sp1 is involved in the regulation of Pol III gene transcription independently of FLNA expression. Reporter gene assays showed that alteration of Sp1 expression affects BRF1P4 and GTF3C2P2 activation, suggesting that Sp1 modulates Pol III-mediated gene transcription by controlling BRF1 and GTF3C2 gene expression. Further analysis revealed that Sp1 interacts with and thereby promotes the occupancies of TATA box-binding protein, TFIIAα, and p300 at both BRF1P4 and GTF3C2P2. These findings indicate that Sp1 controls Pol III-directed transcription and shed light on how Sp1 regulates cancer cell proliferation.


Subject(s)
RNA Polymerase III/metabolism , Sp1 Transcription Factor/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factors, TFIII/metabolism , Binding Sites , Cell Line , Cell Proliferation , E1A-Associated p300 Protein/metabolism , Filamins/antagonists & inhibitors , Filamins/genetics , Filamins/metabolism , Humans , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA Interference , RNA Polymerase III/genetics , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , TATA-Binding Protein Associated Factors/antagonists & inhibitors , TATA-Binding Protein Associated Factors/genetics , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factors, TFIII/antagonists & inhibitors , Transcription Factors, TFIII/genetics , Transcription, Genetic , Up-Regulation
2.
Circulation ; 140(1): 67-79, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31014088

ABSTRACT

BACKGROUND: The actin-binding protein FLNA (filamin A) regulates signal transduction important for cell locomotion, but the role of macrophage-specific FLNA during atherogenesis has not been explored. METHODS: We analyzed FLNA expression in human carotid atherosclerotic plaques by immunofluorescence. We also produced mice with Flna-deficient macrophages by breeding conditional Flna-knockout mice ( Flna o/fl) with mice expressing Cre from the macrophage-specific lysosome M promoter ( LC). Atherosclerosis in vivo was studied by transplanting bone marrow from male Flna o/fl/ LC mice to atherogenic low-density lipoprotein receptor-deficient ( Ldlr-/-) mice; and by infecting Flna o/fl and Flna o/fl/ LC mice with AdPCSK9 (adenoviral vector overexpressing proprotein convertase subtilisin/kexin type 9). Furthermore, C57BL/6 mice were infected with AdPCSK9 and then treated with the calpain inhibitor calpeptin to inhibit FLNA cleavage. RESULTS: We found that macrophage FLNA expression was higher in advanced than in intermediate human atherosclerotic plaques. Flna o/fl/ LC macrophages proliferated and migrated less than controls; expressed lower levels of phosphorylated AKT and ERK1/2; exhibited reduced foam cell formation and lipid uptake; and excreted more lipids. The deficiency of Flna in macrophages markedly reduced the size of aortic atherosclerotic plaques in both Ldlr-/-BMT: Flnao/fl/LC and AdPCSK9-infected Flna o/fl/ LC mice. Intima/media ratios and numbers of CD68-positive macrophages in atherosclerotic plaques were lower in Flna-deficient mice than in control mice. Moreover, we found that STAT3 interacts with a calpain-cleaved carboxyl-terminal fragment of FLNA. Inhibiting calpain-mediated FLNA cleavage with calpeptin in macrophages reduced nuclear levels of phosphorylated STAT3, interleukin 6 secretion, foam cell formation, and lipid uptake. Finally, calpeptin treatment reduced the size of atherosclerotic plaques in C57BL/6 mice infected with AdPCSK9. CONCLUSIONS: Genetic inactivation of Flna and chemical inhibition of calpain-dependent cleavage of FLNA impaired macrophage signaling and function, and reduced atherosclerosis in mice, suggesting that drugs targeting FLNA may be useful in the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/metabolism , Filamins/deficiency , Filamins/genetics , Gene Targeting/methods , Macrophage Activation/physiology , Animals , Atherosclerosis/pathology , Cells, Cultured , Filamins/antagonists & inhibitors , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Cell Rep ; 14(11): 2653-67, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26972007

ABSTRACT

Neuronal function is highly sensitive to changes in oxygen levels, but how hypoxia affects dendritic spine formation and synaptogenesis is unknown. Here we report that hypoxia, chemical inhibition of the oxygen-sensing prolyl hydroxylase domain proteins (PHDs), and silencing of Phd2 induce immature filopodium-like dendritic protrusions, promote spine regression, reduce synaptic density, and decrease the frequency of spontaneous action potentials independently of HIF signaling. We identified the actin cross-linker filamin A (FLNA) as a target of PHD2 mediating these effects. In normoxia, PHD2 hydroxylates the proline residues P2309 and P2316 in FLNA, leading to von Hippel-Lindau (VHL)-mediated ubiquitination and proteasomal degradation. In hypoxia, PHD2 inactivation rapidly upregulates FLNA protein levels because of blockage of its proteasomal degradation. FLNA upregulation induces more immature spines, whereas Flna silencing rescues the immature spine phenotype induced by PHD2 inhibition.


Subject(s)
Dendritic Spines/metabolism , Filamins/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Synapses/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Filamins/antagonists & inhibitors , Filamins/genetics , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Mice , Mice, Knockout , Oxygen/metabolism , Rats , Rats, Wistar , Tubulin/metabolism , Up-Regulation/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
4.
Cell Struct Funct ; 40(2): 61-7, 2015.
Article in English | MEDLINE | ID: mdl-25925610

ABSTRACT

Numerous types of cancer cells migrate into extracellular tissues. This phenomenon is termed invasion, and is associated with poor prognosis in cancer patients. In this study, we demonstrated that filamin B (FLNb), an actin-binding protein, is highly expressed in cancer cell lines that exhibit high invasiveness, with a spindle morphology, into 3D collagen matrices. In addition, we determined that knockdown of FLNb in invasive cancer cells converts cell morphology from spindle-shaped, which is associated with high invasiveness, to round-shaped with low invasiveness. Furthermore, di-phosphorylation of myosin regulatory light chain (MRLC) and phosphorylation of focal adhesion kinase (FAK) are inhibited in FLNb-knockdown cancer cells. These results suggest that FLNb enhances invasion of cancer cells through phosphorylation of MRLC and FAK. Therefore, FLNb may be a new therapeutic target for invasive cancers.


Subject(s)
Epithelial Cells/metabolism , Fibroblasts/metabolism , Filamins/genetics , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Myosin Light Chains/genetics , Cell Adhesion , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Collagen/chemistry , Epithelial Cells/pathology , Fibroblasts/pathology , Filamins/antagonists & inhibitors , Filamins/metabolism , Focal Adhesion Kinase 1/metabolism , Humans , Myosin Light Chains/metabolism , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
5.
PLoS One ; 10(4): e0123018, 2015.
Article in English | MEDLINE | ID: mdl-25861040

ABSTRACT

Polycystin-2 (PC2), encoded by the PKD2 gene, is mutated in ~15% of autosomal dominant polycystic kidney disease. Filamins are actin-binding proteins implicated in scaffolding and membrane stabilization. Here we studied the effects of filamin on PC2 stability using filamin-deficient human melanoma M2, filamin-A (FLNA)-replete A7, HEK293 and IMCD cells together with FLNA siRNA/shRNA knockdown (KD). We found that the presence of FLNA is associated with higher total and plasma membrane PC2 protein expression. Western blotting analysis in combination with FLNA KD showed that FLNA in A7 cells represses PC2 degradation, prolonging the half-life from 2.3 to 4.4 hours. By co-immunoprecipitation and Far Western blotting we found that the FLNA C-terminus (FLNAC) reduces the FLNA-PC2 binding and PC2 expression, presumably through competing with FLNA for binding PC2. We further found that FLNA mediates PC2 binding with actin through forming complex PC2-FLNA-actin. FLNAC acted as a blocking peptide and disrupted the link of PC2 with actin through disrupting the PC2-FLNA-actin complex. Finally, we demonstrated that the physical interaction of PC2-FLNA is Ca-dependent. Taken together, our current study indicates that FLNA anchors PC2 to the actin cytoskeleton through complex PC2-FLNA-actin to reduce degradation and increase stability, and possibly regulate PC2 function in a Ca-dependent manner.


Subject(s)
Filamins/metabolism , TRPP Cation Channels/metabolism , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Filamins/antagonists & inhibitors , Filamins/genetics , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TRPP Cation Channels/chemistry , TRPP Cation Channels/genetics
6.
Cell Rep ; 8(6): 1943-1956, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25242330

ABSTRACT

Colorectal cancer (CRC) initiation and growth is often attributed to stem cells, yet little is known about the regulation of these cells. We show here that a subpopulation of Prox1-transcription-factor-expressing cells have stem cell activity in intestinal adenomas, but not in the normal intestine. Using in vivo models and 3D ex vivo organoid cultures of mouse adenomas and human CRC, we found that Prox1 deletion reduced the number of stem cells and cell proliferation and decreased intestinal tumor growth via induction of annexin A1 and reduction of the actin-binding protein filamin A, which has been implicated as a prognostic marker in CRC. Loss of Prox1 also decreased autophagy and the survival of hypoxic tumor cells in tumor transplants. Thus, Prox1 is essential for the expansion of the stem cell pool in intestinal adenomas and CRC without being critical for the normal functions of the gut.


Subject(s)
Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Annexin A1/antagonists & inhibitors , Annexin A1/genetics , Annexin A1/metabolism , Autophagy , Cell Culture Techniques , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Filamins/antagonists & inhibitors , Filamins/genetics , Filamins/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Ischemia/pathology , Ischemia/prevention & control , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Up-Regulation
7.
Nat Commun ; 5: 4428, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25079702

ABSTRACT

Recent studies have shown that certain types of transformed cells are extruded from an epithelial monolayer. However, it is not known whether and how neighbouring normal cells play an active role in this process. In this study, we demonstrate that filamin A and vimentin accumulate in normal cells specifically at the interface with Src- or RasV12-transformed cells. Knockdown of filamin A or vimentin in normal cells profoundly suppresses apical extrusion of the neighbouring transformed cells. In addition, we show in zebrafish embryos that filamin plays a positive role in the elimination of the transformed cells. Furthermore, the Rho/Rho kinase pathway regulates filamin accumulation and filamin acts upstream of vimentin in the apical extrusion. This is the first report demonstrating that normal epithelial cells recognize and actively eliminate neighbouring transformed cells and that filamin is a key mediator in the interaction between normal and transformed epithelial cells.


Subject(s)
Filamins/genetics , Gene Expression Regulation , Vimentin/genetics , Zebrafish/genetics , Animals , Cell Death , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Dogs , Embryo, Nonmammalian , Filamins/antagonists & inhibitors , Filamins/metabolism , Madin Darby Canine Kidney Cells , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transformation, Genetic , Vimentin/antagonists & inhibitors , Vimentin/metabolism , Zebrafish/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
8.
Dev Biol ; 347(2): 247-57, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20707996

ABSTRACT

Filamin, known primarily for its actin cross-linking function, is a stretch-sensitive structural and signaling scaffold that binds transmembrane receptors and a wide variety of intracellular signaling proteins. The Caenorhabditis elegans filamin ortholog, FLN-1, has a well conserved overall structure, including an N-terminal actin-binding domain, and a series of 20 immunoglobulin (Ig)-like repeats. FLN-1 partially colocalizes with actin filaments in spermathecal and uterine cells. Analysis of phenotypes resulting from a deletion allele and RNAi depletion indicates FLN-1 is required to maintain the actin cytoskeleton in the spermatheca and uterus, and to allow the exit of embryos from the spermatheca. FLN-1 deficient animals accumulate embryos in the spermatheca, lay damaged and unfertilized eggs, and consequently exhibit dramatically reduced brood sizes. The phospholipase PLC-1 is also required for the exit of embryos from the spermatheca, and analysis of doubly mutant animals suggests that PLC-1 and FLN-1 act in the same pathway to promote proper transit of embryos from the spermatheca to the uterus. Given the modular protein structure, subcellular localization, genetic interaction with PLC-1, and known mechanosensory functions of filamin, we postulate that FLN-1 may be required to convert mechanical information about the presence of the oocyte into a biochemical signal, thereby allowing timely exit of the embryo from the spermatheca.


Subject(s)
Actins/metabolism , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Contractile Proteins/physiology , Filamins/physiology , Microfilament Proteins/physiology , Oocytes/physiology , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Contractile Proteins/antagonists & inhibitors , Contractile Proteins/chemistry , Contractile Proteins/genetics , Female , Fertility/genetics , Fertility/physiology , Filamins/antagonists & inhibitors , Filamins/chemistry , Filamins/genetics , Genitalia/growth & development , Genitalia/physiology , Male , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA Interference , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...