Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Sci Rep ; 9(1): 4771, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30886324

ABSTRACT

Nuclear localization signals are short amino acid sequences that target proteins for nuclear import. In this manuscript, we have generated a chimeric tri-functional peptide composed of a cell penetrating peptide (CPP), a nuclear localization sequence and an interfering peptide blocking the interaction between TEAD and YAP, two transcription factors involved in the Hippo signalling pathway, whose deregulation is related to several types of cancer. We have validated the cell penetration and nuclear localization by flow cytometry and fluorescence microscopy and shown that the new generated peptide displays an apoptotic effect in tumor cell lines thanks to the specific nuclear delivery of the cargo, which targets a protein/protein interaction in the nucleus. In addition, the peptide has an anti-tumoral effect in vivo in xenograft models of breast cancer. The chimeric peptide designed in the current study shows encouraging prospects for developing nuclear anti- neoplastic drugs.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Breast Neoplasms/drug therapy , DNA-Binding Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Peptides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Drug Delivery Systems , Female , Hippo Signaling Pathway , Humans , Male , Mice , Mice, Inbred C3H , Nuclear Localization Signals/metabolism , Nuclear Proteins/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , TEA Domain Transcription Factors , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
2.
Anal Chem ; 90(22): 13356-13364, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30234969

ABSTRACT

Metallic plasmonic nanoparticles have been intensively exploited as theranostic nanoprobes for plasmonic photothermal therapy (PPT) and surface-enhanced Raman spectroscopy (SERS) applications. But the underlying molecular mechanisms associated with PPT-induced apoptosis between cancerous and normal cells have remained largely unknown or disputed. In this study, we designed an organelle-targeting theranostic plasmonic SERS nanoprobe (CDs-Ag/Au NS) composed of porous Ag/Au nanoshell (p-Ag/Au NSs) and carbon dots (CDs) for nucleus and mitochondria targeted PPT of cells. The differences in molecular stress response in the PPT-induced hyperthermia cell death between cancerous HeLa and normal L929 and H8 cells have been revealed by site-specific single-cell SERS detection. The contents of tryptophan (Trp), phenylalanine (Phe), and tyrosine (Tyr) in HeLa cells were found more evidently increased than L929 and H8 cells during the PPT-induced cell-death process. And from the mitochondria point of view, we found that the PPT-induced cell apoptosis for HeLa cells mainly stems from (or is regulated through) cellular thermal stress-responsive proteins, while for L929 and H8 cells it seems more related to DNA. Understanding molecular stress response difference of the PPT-induced cell apoptosis between cancerous and normal cells is helpful for diagnosis and treatment of cancer, and the method will open an avenue for single-cell studies.


Subject(s)
Cell Nucleus/metabolism , Mitochondria/metabolism , Nanoshells/chemistry , Quantum Dots/chemistry , Spectrum Analysis, Raman/methods , Theranostic Nanomedicine/methods , Apoptosis/drug effects , Carbon/chemistry , Carbon/radiation effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , DNA/metabolism , DNA Fragmentation/drug effects , Gold/chemistry , Gold/radiation effects , HeLa Cells , Humans , Hyperthermia, Induced/methods , Infrared Rays , Nanoshells/radiation effects , Neoplasms/metabolism , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Quantum Dots/radiation effects , Silver/chemistry , Silver/radiation effects
3.
Mol Cell Proteomics ; 12(1): 145-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23087160

ABSTRACT

The human importin-ß family consists of 21 nucleocytoplasmic transport carrier proteins that carry proteins and RNAs across the nuclear envelope through nuclear pores in specific directions. These transport carriers are responsible for the nucleocytoplasmic transport of thousands of proteins, but the cargo allocation of each carrier, which is necessary information if one wishes to understand the physiological context of transport, is poorly characterized. To address this issue, we developed a high-throughput method to identify the cargoes of transport carriers by applying stable isotope labeling by amino acids in cell culture to construct an in vitro transport system. Our method can be outlined in three steps. (1) Cells are cultured in a medium containing a stable isotope. (2) The cell membranes of the labeled cells are permeabilized, and proteins extracted from unlabeled cells are transported into the nuclei of the permeabilized cells. In this step, the reaction system is first depleted of all importin-ß family carriers and then supplemented with a particular importin-ß family carrier of interest. (3) Proteins in the nuclei are extracted and analyzed quantitatively via LC-MS/MS. As an important test case, we used this method to identify cargo proteins of transportin, a representative member of the importin-ß family. As expected, the identified candidate cargo proteins included previously reported transportin cargoes as well as new potential cargoes, which we corroborated via in vitro binding assays. The identified cargoes are predominately RNA-interacting proteins, affirming that cargoes allotted to the same carrier share functional characteristics. Finally, we found that the transportin cargoes possessed at least two classes of signal sequences: the well characterized PY-nuclear localization signals specific for transportin, and Lys/Arg-rich segments capable of binding to both transportin and importin-ß. Thus, our method will be useful for linking a carrier to features shared among its cargoes and to specific nuclear localization signals.


Subject(s)
Active Transport, Cell Nucleus , Nuclear Localization Signals/metabolism , beta Karyopherins/analysis , Amino Acid Sequence , Amino Acids , Cell Membrane , Chromatography, Liquid , Humans , Isotope Labeling , Nuclear Envelope/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proteomics , Tandem Mass Spectrometry , beta Karyopherins/metabolism
4.
Development ; 136(16): 2767-77, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19633170

ABSTRACT

During neurogenesis in Xenopus, apicobasally polarised superficial and non-polar deep cells take up different fates: deep cells become primary neurons while superficial cells stay as progenitors. It is not known whether the proteins that affect cell polarity also affect cell fate and how membrane polarity information may be transmitted to the nucleus. Here, we examine the role of the polarity components, apically enriched aPKC and basolateral Lgl2, in primary neurogenesis. We report that a membrane-tethered form of aPKC (aPKC-CAAX) suppresses primary neurogenesis and promotes cell proliferation. Unexpectedly, both endogenous aPKC and aPKC-CAAX show some nuclear localisation. A constitutively active aPKC fused to a nuclear localisation signal has the same phenotypic effect as aPKC-CAAX in that it suppresses neurogenesis and enhances proliferation. Conversely, inhibiting endogenous aPKC with a dominant-negative form that is restricted to the nucleus enhances primary neurogenesis. These observations suggest that aPKC has a function in the nucleus that is important for cell fate specification during primary neurogenesis. In a complementary experiment, overexpressing basolateral Lgl2 causes depolarisation and internalisation of superficial cells, which form ectopic neurons when supplemented with a proneural factor. These findings suggest that both aPKC and Lgl2 affect cell fate, but that aPKC is a nuclear determinant itself that might shuttle from the membrane to the nucleus to control cell proliferation and fate; loss of epithelial cell polarity by Lgl2 overexpression changes the position of the cells and is permissive for a change in cell fate.


Subject(s)
Cell Nucleus/physiology , Cell Proliferation , Neurogenesis/physiology , Protein Kinase C/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , beta Karyopherins/metabolism , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Polarity , HeLa Cells , Humans , In Situ Hybridization , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Tubulin/metabolism , Xenopus Proteins/genetics , beta Karyopherins/genetics
5.
Chembiochem ; 7(1): 88-97, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16345111

ABSTRACT

Nuclear import triggered by the nuclear-localisation sequence (NLS) of the viral Jun (vJun) protein is mediated by phosphorylation of a serine close to the NLS. Since phosphorylation and glycosylation of serine residues are often in a reciprocal "yin-yang" relationship, we investigated whether glycosylation of this serine with O-linked N-acetylglucosamine (O-GlcNAc) would also regulate nuclear import via the vJun NLS. Peptides containing the vJun NLS with an adjacent O-phosphorylated, O-GlcNAc-functionalised or unmodified serine, and equipped with an N-terminal biotin or a 7-nitrobenz-2-oxa-1,3-diazolyl (NBD) fluorescent label, were synthesised on the solid phase by means of an Fmoc/Boc strategy and a Pd0-sensitive HYCRON linker. Fluorescence-polarisation measurements on the NBD-labelled peptides indicated that modification with phosphate or O-GlcNAc leads to a decrease in affinity to the import-mediating adapter protein, importin alpha, of about one order of magnitude compared to the unmodified NLS. Microinjection of biotinylated NLS peptide conjugated with fluorescently labelled avidin into NIH/3T3 and MDCK cells, revealed that avidin-unmodified-NLS peptide was rapidly imported into the nucleus. However, either phosphate or O-GlcNAc next to the NLS caused almost complete exclusion of the protein conjugate from nuclear import. These findings indicate that nuclear import by the vJun NLS might not be regulated by a "yin-yang" modification of an adjacent serine with phosphate or O-GlcNAc. Rather, negative regulation of binding between the polybasic NLS and importin by a negatively charged or a bulky, uncharged residue appears likely.


Subject(s)
Cell Nucleus/metabolism , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Oncogene Protein p65(gag-jun)/chemistry , Serine/chemistry , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Avidin/chemistry , Avidin/metabolism , Cell Nucleus/chemistry , Cells, Cultured , Dogs , Glycosylation , Mice , Molecular Structure , NIH 3T3 Cells , Nuclear Localization Signals/chemical synthesis , Phosphorylation , Serine/metabolism , Structure-Activity Relationship
6.
Cell Res ; 15(7): 504-10, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16045813

ABSTRACT

ErbB2, a member of the receptor tyrosine kinase family, is frequently over-expressed in breast cancer. Proteolysis of the extracellular domain of ErbB2 results in constitutive activation of ErbB2 kinase. Recent study reported that ErbB2 is found in the nucleus. Here, we showed that ErbB2 is imported into the nucleus through a nuclear localization signal (NLS)-mediated mechanism. The NLS sequence KRRQQKIRKYTMRR (aa655-668) contains three clusters of basic amino acids and it is sufficient to target GFP into the nucleus. However, mutation in any basic amino acid cluster of this NLS sequence significantly affects its nuclear localization. Furthermore, it was found that this NLS is essential for the nuclear localization of ErbB2 since the intracellular domain of Erb2 lacking NLS completely abrogates its nuclear translocation. Taken together, our study identified a novel nuclear localization signal and reveals a novel mechanism underlying ErbB2 nuclear trafficking and localization.


Subject(s)
Nuclear Localization Signals/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Animals , COS Cells , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation , Humans , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary/physiology , Receptor, ErbB-2/genetics
8.
Biochem J ; 361(Pt 3): 505-14, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11802780

ABSTRACT

5-Lipoxygenase (5-LO) metabolizes arachidonic acid to leukotriene A4, a key intermediate in leukotriene biosynthesis. To explore the molecular mechanisms of its cell-specific localization, a fusion protein between green fluorescent protein (GFP) and human 5-LO (GFP-5LO) was expressed in various cells. GFP-5LO was localized in the cytosol in HL-60 cells and in both the nucleus and the cytosol in RBL (rat basophilic leukaemia) cells, similarly to the native enzyme in these cells. The localization of GFP fusion proteins for mutant 5-LOs in a putative bipartite nuclear localization signal (NLS), amino acids 638-655, in Chinese hamster ovary (CHO)-K1 and Swiss3T3 cells revealed that this motif is important for the nuclear localization of 5-LO. A GFP fusion protein of this short peptide localized consistently in the nucleus. Leptomycin B, a specific inhibitor of nuclear export signal (NES)-dependent transport, diminished the cytosolic localization of 5-LO in HL-60 cells and that of GFP-5LO in CHO-K1 cells, suggesting that an NES-system might also function in determining 5-LO localization. Analysis of the localization of 5-LO during the cell cycle points to a controlled movement of this enzyme. Thus we conclude that a balance of NLS- and NES-dependent mechanisms determines the cell-type-specific localization of 5-LO, suggesting a nuclear function for this enzyme.


Subject(s)
Arachidonate 5-Lipoxygenase/biosynthesis , Cell Nucleus/metabolism , Nuclear Localization Signals/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Sequence , Animals , Arachidonate 5-Lipoxygenase/chemistry , CHO Cells , Cell Cycle , Cell Line , Chromatography, High Pressure Liquid , Cricetinae , Cytosol/metabolism , DNA, Complementary/metabolism , Fatty Acids, Unsaturated/pharmacology , Green Fluorescent Proteins , HL-60 Cells , Humans , Immunoblotting , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Protein Binding , Rats , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
9.
Plant Physiol ; 124(3): 979-90, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11080276

ABSTRACT

The Arabidopsis COP1 protein functions as a developmental regulator, in part by repressing photomorphogenesis in darkness. Using complementation of a cop1 loss-of-function allele with transgenes expressing fusions of cop1 mutant proteins and beta-glucuronidase, it was confirmed that COP1 consists of two modules, an amino terminal module conferring a basal function during development and a carboxyl terminal module conferring repression of photomorphogenesis. The amino-terminal zinc-binding domain of COP1 was indispensable for COP1 function. In contrast, the debilitating effects of site-directed mutations in the single nuclear localization signal of COP1 were partially compensated by high-level transgene expression. The carboxyl-terminal module of COP1, though unable to substantially ameliorate a cop1 loss-of-function allele on its own, was sufficient for conferring a light-quality-dependent hyperetiolation phenotype in the presence of wild-type COP1. Moreover, partial COP1 activity could be reconstituted in vivo from two non-covalently linked, complementary polypeptides that represent the two functional modules of COP1. Evidence is presented for efficient association of the two sub-fragments of the split COP1 protein in Arabidopsis and in a yeast two-hybrid assay.


Subject(s)
Arabidopsis Proteins , Arabidopsis/genetics , Carrier Proteins/genetics , Nuclear Localization Signals/genetics , Plant Proteins/genetics , Repressor Proteins/genetics , Ubiquitin-Protein Ligases , Arabidopsis/chemistry , Arabidopsis/metabolism , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Genetic Complementation Test , Glucuronidase/genetics , Glucuronidase/metabolism , Light , Mutagenesis, Site-Directed , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phenotype , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Structures/genetics , Plant Structures/growth & development , Plant Structures/metabolism , Plants, Genetically Modified , Precipitin Tests , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Two-Hybrid System Techniques
10.
Nucleic Acids Symp Ser ; (44): 265-6, 2000.
Article in English | MEDLINE | ID: mdl-12903370

ABSTRACT

Transcription factor Sp1 is located in the nucleus of a mammalian cell and importantly related to expression of many cellular genes. In order to elucidate the nuclear localization mechanism of Sp1, various truncated fragments of Sp1 were fused to green fluorescent protein (GFP) and expressed in HeLa cell. The results show significance of the DNA binding region, especially, zinc finger (Zn finger) domain for nuclear localization of Sp1 in HeLa cell.


Subject(s)
Podophyllin/analogs & derivatives , Podophyllin/chemistry , Podophyllin/metabolism , Binding Sites , Cell Nucleus/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Structure , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Podophyllin/genetics , Podophyllotoxin/analogs & derivatives , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL