Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Opin Cell Biol ; 68: 173-180, 2021 02.
Article in English | MEDLINE | ID: mdl-33227657

ABSTRACT

The nucleoskeleton has been associated with partitioning the genome into active and inactive compartments that dictate local transcription factor (TF) activity. However, recent data indicate that the nucleoskeleton and TFs reciprocally influence each other in dynamic TF trafficking pathways through the functions of LEM proteins. While the conserved peripheral recruitment of TFs by LEM proteins has been viewed as a mechanism of repressing transcription, a diversity of release mechanisms from the lamina suggest this compartment serves as a refuge for nuclear TF accumulation for rapid mobilization and signal stability. Detailed mechanisms suggest that TFs toggle between nuclear lamina refuge and nuclear matrix lamin-LEM protein complexes at sites of active transcription. In this review we will highlight emerging LEM functions acting at the interface of chromatin and nucleoskeleton to create TF trafficking networks.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Humans , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism
2.
Cell ; 176(1-2): 198-212.e15, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30503211

ABSTRACT

Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2ß forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2ß for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.


Subject(s)
DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Zinc Finger Protein GLI1/metabolism , 3T3 Cells , Animals , Carcinoma, Basal Cell/metabolism , Cell Line , Chromatin , DNA-Binding Proteins/physiology , HEK293 Cells , Hedgehog Proteins/metabolism , Hedgehog Proteins/physiology , Histone Deacetylase 1/metabolism , Humans , Membrane Proteins/physiology , Mice , Molecular Chaperones/metabolism , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1/physiology , Zinc Fingers
3.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093271

ABSTRACT

Advanced basal cell carcinomas (BCCs) circumvent Smoothened (SMO) inhibition by activating GLI transcription factors to sustain the high levels of Hedgehog (HH) signaling required for their survival. Unfortunately, there is a lack of efficacious therapies. We performed a gene expression-based drug repositioning screen in silico and identified the FDA-approved histone deacetylase (HDAC) inhibitor, vorinostat, as a top therapeutic candidate. We show that vorinostat only inhibits proliferation of BCC cells in vitro and BCC allografts in vivo at high dose, limiting its usefulness as a monotherapy. We leveraged this in silico approach to identify drug combinations that increase the therapeutic window of vorinostat and identified atypical PKC Ɩ/ʎ (aPKC) as a HDAC costimulator of HH signaling. We found that aPKC promotes GLI1-HDAC1 association in vitro, linking two positive feedback loops. Combination targeting of HDAC1 and aPKC robustly inhibited GLI1, lowering drug doses needed in vitro, in vivo, and ex vivo in patient-derived BCC explants. We identified a bioavailable and selective small-molecule aPKC inhibitor, bringing the pharmacological blockade of aPKC and HDAC1 into the realm of clinical possibility. Our findings provide a compelling rationale and candidate drugs for combined targeting of HDAC1 and aPKC in HH-dependent cancers.


Subject(s)
Carcinoma, Basal Cell/drug therapy , Histone Deacetylase 1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Isoenzymes/drug effects , Protein Kinase C/drug effects , Skin Neoplasms/drug therapy , Allografts , Animals , Carcinoma, Basal Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology , Drug Combinations , Drug Discovery , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hedgehogs/genetics , Hedgehogs/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemistry , Isoenzymes/metabolism , Mice , Mice, Knockout , Protein Kinase C/metabolism , Signal Transduction , Transcription Factors/drug effects , Transcription Factors/genetics , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...