Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32453403

ABSTRACT

The mechanisms underlying turnover of the nuclear pore complex (NPC) and the component nucleoporins (Nups) are still poorly understood. In this study, we found that the budding yeast Saccharomyces cerevisiae triggers NPC degradation by autophagy upon the inactivation of Tor kinase complex 1. This degradation largely depends on the selective autophagy-specific factor Atg11 and the autophagy receptor-binding ability of Atg8, suggesting that the NPC is degraded via receptor-dependent selective autophagy. Immunoelectron microscopy revealed that NPCs embedded in nuclear envelope-derived double-membrane vesicles are sequestered within autophagosomes. At least two pathways are involved in NPC degradation: Atg39-dependent nucleophagy (selective autophagy of the nucleus) and a pathway involving an unknown receptor. In addition, we found the interaction between Nup159 and Atg8 via the Atg8-family interacting motif is important for degradation of this nucleoporin not assembled into the NPC. Thus, this study provides the first evidence for autophagic degradation of the NPC and Nups, which we term "NPC-phagy" and "nucleoporinophagy."


Subject(s)
Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Proteins/genetics , Autophagy/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Vesicular Transport Proteins/genetics , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Proteins/metabolism , Gene Expression Regulation, Fungal , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Microscopy, Immunoelectron , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Sirolimus/pharmacology , Vesicular Transport Proteins/metabolism
2.
Nat Commun ; 10(1): 3827, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31444357

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of unknown etiology. Although defects in nucleocytoplasmic transport (NCT) may be central to the pathogenesis of ALS and other neurodegenerative diseases, the molecular mechanisms modulating the nuclear pore function are still largely unknown. Here we show that genetic and pharmacological modulation of actin polymerization disrupts nuclear pore integrity, nuclear import, and downstream pathways such as mRNA post-transcriptional regulation. Importantly, we demonstrate that modulation of actin homeostasis can rescue nuclear pore instability and dysfunction caused by mutant PFN1 as well as by C9ORF72 repeat expansion, the most common mutation in ALS patients. Collectively, our data link NCT defects to ALS-associated cellular pathology and propose the regulation of actin homeostasis as a novel therapeutic strategy for ALS and other neurodegenerative diseases.


Subject(s)
Actins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Nuclear Pore/pathology , Profilins/metabolism , Acrylamides/pharmacology , Actins/ultrastructure , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Amyotrophic Lateral Sclerosis/genetics , Biopsy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Embryo, Mammalian , Fibroblasts , Humans , Microscopy, Electron, Transmission , Motor Neurons/cytology , Mutation , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Primary Cell Culture , Profilins/genetics , Protein Multimerization/drug effects , Protein Multimerization/genetics , Skin/cytology , Skin/pathology , Thiazoles/pharmacology , Thiazolidines/pharmacology
3.
ACS Nano ; 11(6): 5567-5578, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28530826

ABSTRACT

Nuclear pore complexes (NPCs) are the sole turnstile implanted in the nuclear envelope (NE), acting as a central nanoregulator of transport between the cytosol and the nucleus. NPCs consist of ∼30 proteins, termed nucleoporins. About one-third of nucleoporins harbor natively unstructured, intrinsically disordered phenylalanine-glycine strings (FG-Nups), which engage in transport selectivity. Because the barriers insert deeply in the NPC, they are nearly inaccessible. Several in vitro barrier models have been proposed; however, the dynamic FG-Nups protein molecules themselves are imperceptible in vivo. We show here that high-speed atomic force microscopy (HS-AFM) can be used to directly visualize nanotopographical changes of the nuclear pore inner channel in colorectal cancer (CRC) cells. Furthermore, using MLN8237/alisertib, an apoptotic and autophagic inducer currently being tested in relapsed cancer clinical trials, we unveiled the functional loss of nucleoporins, particularly the deformation of the FG-Nups barrier, in dying cancer cells. We propose that the loss of this nanoscopic resilience is an irreversible dying code in cells. These findings not only illuminate the potential application of HS-AFM as an intracellular nanoendoscopy but also might aid in the design of future nuclear targeted nanodrug delivery tailored to the individual patient.


Subject(s)
Colorectal Neoplasms/pathology , Microscopy, Atomic Force/methods , Nuclear Pore/pathology , Apoptosis/drug effects , Azepines/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , HCT116 Cells , Humans , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/analysis , Nuclear Pore Complex Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
4.
Proc Natl Acad Sci U S A ; 114(7): E1111-E1117, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28069952

ABSTRACT

The toxic proline:arginine (PRn) poly-dipeptide encoded by the (GGGGCC)n repeat expansion in the C9orf72 form of heritable amyotrophic lateral sclerosis (ALS) binds to the central channel of the nuclear pore and inhibits the movement of macromolecules into and out of the nucleus. The PRn poly-dipeptide binds to polymeric forms of the phenylalanine:glycine (FG) repeat domain, which is shared by several proteins of the nuclear pore complex, including those in the central channel. A method of chemical footprinting was used to characterize labile, cross-ß polymers formed from the FG domain of the Nup54 protein. Mutations within the footprinted region of Nup54 polymers blocked both polymerization and binding by the PRn poly-dipeptide. The aliphatic alcohol 1,6-hexanediol melted FG domain polymers in vitro and reversed PRn-mediated enhancement of the nuclear pore permeability barrier. These data suggest that toxicity of the PRn poly-dipeptide results in part from its ability to lock the FG repeats of nuclear pore proteins in the polymerized state. Our study offers a mechanistic interpretation of PRn poly-dipeptide toxicity in the context of a prominent form of ALS.


Subject(s)
Active Transport, Cell Nucleus , C9orf72 Protein/pharmacology , DNA Repeat Expansion/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Biopolymers , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/genetics , Dipeptides/metabolism , Dipeptides/pharmacology , Female , Glycols/pharmacology , Humans , Microscopy, Confocal , Nuclear Pore/chemistry , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/ultrastructure , Oocytes/ultrastructure , Permeability/drug effects , Protein Binding , Protein Domains , Wheat Germ Agglutinins/metabolism , Wheat Germ Agglutinins/pharmacology , Xenopus laevis
5.
Apoptosis ; 22(3): 393-405, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28000054

ABSTRACT

Regulation of nuclear transport is an essential component of apoptosis. As chemotherapy induced cell death progresses, nuclear transport and the nuclear pore complex (NPC) are slowly disrupted and dismantled. 5-Fluorouracil (5-FU) and the camptothecin derivatives irinotecan and topotecan, are linked to altered nuclear transport of specific proteins; however, their general effects on the NPC and transport during apoptosis have not been characterized. We demonstrate that 5-FU, but not topotecan, increases NPC permeability, and disrupts Ran-mediated nuclear transport before the disruption of the NPC. This increased permeability is dependent on increased cellular calcium, as the Ca2+ chelator BAPTA-AM, abolishes the effect. Furthermore, increased calcium alone was sufficient to disrupt the Ran gradient. Combination treatments of 5-FU with topotecan or irinotecan, similarly disrupted nuclear transport before disassembly of the NPC. In both single and combination treatments nuclear transport was disrupted before caspase 9 activation, indicating that 5-FU induces an early caspase-independent increase in NPC permeability and alteration of nuclear transport. Because Crm1-mediated nuclear export of tumor suppressors is linked to drug resistance we also examined the effect of 5-FU on the nuclear export of a specific target, topoisomerase. 5-FU treatment led to accumulation of topoisomerase in the nucleus and recovered the loss nuclear topoisomerase induced by irinotecan or topotecan, a known cause of drug resistance. Furthermore, 5-FU retains its ability to cause nuclear accumulation of p53 in the presence of irinotecan or topotecan. Our results reveal a new mechanism of action for these therapeutics during apoptosis, opening the door to other potential combination chemotherapies that employ 5-FU as a calcium mediated inhibitor of Crm1-induced nuclear export of tumor suppressors.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Calcium/physiology , Fluorouracil/pharmacology , Nuclear Pore/drug effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Caspases/metabolism , Cell Nucleus/enzymology , DNA Topoisomerases, Type I/metabolism , Drug Interactions , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , HeLa Cells , Humans , Irinotecan , Neoplasm Proteins/physiology , Permeability , Topotecan/pharmacology , Tumor Suppressor Protein p53/metabolism , ran GTP-Binding Protein/physiology
7.
PLoS One ; 10(4): e0122684, 2015.
Article in English | MEDLINE | ID: mdl-25853427

ABSTRACT

A role for type A Clostridium perfringens in acute hemorrhagic and necrotizing gastroenteritis in dogs and in necrotizing enterocolitis of neonatal foals has long been suspected but incompletely characterized. The supernatants of an isolate made from a dog and from a foal that died from these diseases were both found to be highly cytotoxic for an equine ovarian (EO) cell line. Partial genome sequencing of the canine isolate revealed three novel putative toxin genes encoding proteins related to the pore-forming Leukocidin/Hemolysin Superfamily; these were designated netE, netF, and netG. netE and netF were located on one large conjugative plasmid, and netG was located with a cpe enterotoxin gene on a second large conjugative plasmid. Mutation and complementation showed that only netF was associated with the cytotoxicity. Although netE and netG were not associated with cytotoxicity, immunoblotting with specific antisera showed these proteins to be expressed in vitro. There was a highly significant association between the presence of netF with type A strains isolated from cases of canine acute hemorrhagic gastroenteritis and foal necrotizing enterocolitis. netE and netF were found in all cytotoxic isolates, as was cpe, but netG was less consistently present. Pulsed-field gel electrophoresis showed that netF-positive isolates belonged to a clonal population; some canine and equine netF-positive isolates were genetically indistinguishable. Equine antisera to recombinant Net proteins showed that only antiserum to rNetF had high supernatant cytotoxin neutralizing activity. The identifica-tion of this novel necrotizing toxin is an important advance in understanding the virulence of type A C. perfringens in specific enteric disease of animals.


Subject(s)
Bacterial Toxins/genetics , Clostridium perfringens/genetics , Enterocolitis, Necrotizing/microbiology , Enterotoxins/genetics , Gastroenteritis/microbiology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Cell Line/drug effects , Clostridium perfringens/pathogenicity , Dogs , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/veterinary , Enterotoxins/pharmacology , Gastroenteritis/genetics , Gastroenteritis/veterinary , Genome , High-Throughput Nucleotide Sequencing , Horses , Nuclear Pore/drug effects
8.
PLoS One ; 7(8): e42501, 2012.
Article in English | MEDLINE | ID: mdl-22880006

ABSTRACT

NTF2 is a cytosolic protein responsible for nuclear import of Ran, a small Ras-like GTPase involved in a number of critical cellular processes, including cell cycle regulation, chromatin organization during mitosis, reformation of the nuclear envelope following mitosis, and controlling the directionality of nucleocytoplasmic transport. Herein, we provide evidence for the first time that translocation of the mammalian NTF2 from the nucleus to the cytoplasm to collect Ran in the GDP form is subjected to regulation. Treatment of mammalian cells with polysorbitan monolaurate was found to inhibit nuclear export of tRNA and proteins, which are processes dependent on RanGTP in the nucleus, but not nuclear import of proteins. Inhibition of the export processes by polysorbitan monolaurate is specific and reversible, and is caused by accumulation of Ran in the cytoplasm because of a block in translocation of NTF2 to the cytoplasm. Nuclear import of Ran and the nuclear export processes are restored in polysorbitan monolaurate treated cells overproducing NTF2. Moreover, increased phosphorylation of a phospho-tyrosine protein and several phospho-threonine proteins was observed in polysorbitan monolaurate treated cells. Collectively, these findings suggest that nucleocytoplasmic translocation of NTF2 is regulated in mammalian cells, and may involve a tyrosine and/or threonine kinase-dependent signal transduction mechanism(s).


Subject(s)
Cell Nucleus/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Pregnancy Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Cell Nucleus/drug effects , Deoxycholic Acid/pharmacology , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Nuclear Export Signals , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Phosphorylation/drug effects , Phosphothreonine/metabolism , Phosphotyrosine/metabolism , Polysorbates/pharmacology , Protein Transport/drug effects , RNA Transport/drug effects , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , ran GTP-Binding Protein/antagonists & inhibitors
9.
J Gene Med ; 14(7): 491-500, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22711445

ABSTRACT

BACKGROUND: The nuclear membrane of differentiated airway epithelial cells is a significant barrier for nonviral vectors. Trans-cyclohexane-1,2-diol (TCHD) is an amphipathic alcohol that has been shown to collapse nuclear pore cores and allow the uptake of macromolecules that would otherwise be too large for nuclear entry. Previous studies have shown that TCHD can increase lipid-mediated transfection in vitro. METHODS: We aimed to reproduce these in vitro studies using the cationic lipid GL67A, which we are currently assessing in cystic fibrosis trials and, more importantly, we assessed the effects of TCHD on transfection efficiency in differentiated airway epithelium ex vivo and in mouse lung in vivo using three different drug delivery protocols (nebulisation and bolus administration of TCHD to the mouse lung, as well as perfusion of TCHD to the nasal epithelium, which prolongs contact time between the airway epithelium and drug). RESULTS: TCHD (0.5-2%) dose-dependently increased Lipofectamine 2000 and GL67A-mediated transfection of 293T cells by up to 2 logs. Encouragingly, exposure to 8% TCHD (but not 0.5% or 2.0%) increased gene expression in fully differentiated human air liquid interface cultures by approximately 20-fold, although this was accompanied by significant cell damage. However, none of the TCHD treated mice in any of the three protocols had higher gene expression compared to no TCHD controls. CONCLUSIONS: Although TCHD significantly increases gene transfer in cell lines and differentiated airway epithelium ex vivo, this effect is lost in vivo and further highlights that promising in vitro findings often cannot be translated into in vivo applications.


Subject(s)
Cyclohexanes/pharmacology , Cyclohexanols/pharmacology , Gene Transfer Techniques , Nuclear Pore/drug effects , Respiratory System/drug effects , Animals , Cells, Cultured , Cyclohexanes/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Routes , Epithelium/drug effects , Female , Genetic Therapy , Genetic Vectors , Humans , Lipids/pharmacology , Lung/drug effects , Mice , Mice, Inbred BALB C , Nasal Mucosa/drug effects , Transfection
10.
J Control Release ; 160(3): 601-8, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22386519

ABSTRACT

The efficiency of gene therapy in non-dividing cells is particularly poor due to restricted nuclear delivery rates of exogenously applied macromolecules across the nuclear pore complexes (NPCs). Therefore, improved intranuclear delivery of transgenes requires an ability to modulate the barrier function of the NPC. Despite a large body of experimental evidence accumulated to date, the contribution of individual NPC proteins (nucleoporins) to the formation of the NPC permeability barrier as well as their structural organization within the NPC remains under debate. In the present study, we revisit the view on the spatial arrangement of the Phe-Gly rich domains (FG-domains) of a subset of nucleoporins known as FG-nucleoporins. They are generally believed to be the key constituents of the NPC permeability barrier. Comparison of the binding pattern of a transport receptor importin ß fragment, that binds specifically to FG-domains, with the binding pattern of wheat germ agglutinin that binds elsewhere in the NPC, reveals that FG-domains tend to cluster in the very center of the NPC. Furthermore, a controlled sequential release of the barrier-forming nucleoporins results in a gradual breakdown of the NPC permeability barrier. The breakdown is initiated by a dissociation of Nup62 from the NPC. This is accompanied by an increased passive diffusion of small molecules across the NPC. Subsequent dissociation of Nup98 and possibly other nucleoporins results in a collapse of the barrier for larger molecules. We therefore conclude that FG-nucleoporins do not contribute equally to the maintenance of the NPC permeability barrier exclusion limit. This implies that a controlled release of nucleoporins that contribute most to the formation and maintenance of the NPC barrier can facilitate access of therapeutic macromolecules into the nucleus.


Subject(s)
Nuclear Pore Complex Proteins/chemistry , Nuclear Pore/chemistry , Animals , Cyclohexanols/pharmacology , Female , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Oocytes , Permeability/drug effects , Protein Structure, Tertiary , Xenopus laevis
11.
Anesthesiology ; 116(4): 903-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22314297

ABSTRACT

BACKGROUND: The recent discovery that camphor activates and strongly desensitizes the capsaicin-sensitive and noxious heat-sensitive channel transient receptor potential vanilloid subfamily member 1 (TRPV1) has provided new insights and opened up new research paths toward understanding why this naturally occurring monoterpene is widely used in human medicine for its local counter-irritant, antipruritic, and anesthetic properties. However, the molecular basis for camphor sensitivity remains mostly unknown. The authors attempt to explore the nature of the activation pathways evoked by camphor and narrow down a putative interaction site at TRPV1. METHODS: The authors transiently expressed wild-type or specifically mutated recombinant TRPV1 channels in human embryonic kidney cells HEK293T and recorded cation currents with the whole cell, patch clamp technique. To monitor changes in the spatial distribution of phosphatidylinositol 4,5-bisphosphate, they used fluorescence resonance energy transfer measurements from cells transfected with the fluorescent protein-tagged pleckstrin homology domains of phospholipase C. RESULTS: The results revealed that camphor modulates TRPV1 channel through the outer pore helix domain by affecting its overall gating equilibrium. In addition, camphor, which generally is known to decrease the fluidity of cell plasma membranes, may also regulate the activity of TRPV1 by inducing changes in the spatial distribution of phosphatidylinositol-4,5-bisphosphate on the inner leaflet of the plasma membrane. CONCLUSIONS: The findings of this study provide novel insights into the structural basis for the modulation of TRPV1 channel by camphor and may provide an explanation for the mechanism by which camphor modulates thermal sensation in vivo.


Subject(s)
Camphor/pharmacology , TRPV Cation Channels/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Camphor/chemistry , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Rats , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/chemistry
12.
Am J Pathol ; 180(1): 375-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22074739

ABSTRACT

Phenotypic diversity arises in tumors just as it does in developing organisms, and tumor recurrence frequently manifests from the selective survival of divergent drug-resistant cells. Although the expanding tumor cell population may be successfully targeted, drug-resistant cells may persist and sustain the tumor or enter dormancy before igniting a future relapse. Herein, we show that partial knockdown of nucleoporin p62 (NUP62) by small-interfering RNA confers cisplatin resistance to cultured high-grade ovarian carcinoma cells. Treatment with NUP62 small-interfering RNA and cisplatin leaves resistant cells in a state of dormancy; some dormant cells can be induced to proliferate by transient induction of NUP62 expression from an ectopic expression construct. In addition to suggesting functional links between nuclear pore complex architecture and cancer cell survival, the culture system provides a novel experimental window into the dynamics of tumor cell drug resistance and dormancy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Membrane Glycoproteins/pharmacology , Nuclear Pore Complex Proteins/pharmacology , Nuclear Pore/drug effects , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Female , Gene Knockdown Techniques , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neoplasm Recurrence, Local/genetics , Nuclear Pore/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phenotype , RNA, Small Interfering/genetics
13.
Autophagy ; 7(11): 1392-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21804352

ABSTRACT

We now seem to live in a small world, in which everyone is highly interconnected. Cells, too, often also display tremendous interconnectivities in their component systems. As a recent case in point, we have identified a conserved protein complex--the SEA complex--that links the nuclear pore complex (NPC), the COPII vesicle coating complex, vacuoles and autophagy. In this punctum we will discuss the properties of this novel complex.


Subject(s)
COP-Coated Vesicles/metabolism , Multiprotein Complexes/metabolism , Nitrogen/deficiency , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , COP-Coated Vesicles/drug effects , Multiprotein Complexes/chemistry , Nitrogen/pharmacology , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Protein Binding/drug effects , Protein Structure, Secondary , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Vacuoles/drug effects
14.
Nucleus ; 2(2): 113-8, 2011.
Article in English | MEDLINE | ID: mdl-21738834

ABSTRACT

In eukaryotic cells, the nucleus is a complex and sophisticated organelle containing genomic DNA and supports essential cellular activities. Its surface contains many nuclear pore complexes (NPCs), channels for macromolecular transport between the cytoplasm and nucleus. It has been observed that the nuclear volume and the number of NPCs almost doubles during interphase in dividing cells, but the coordination of these events with the cell cycle was poorly understood, particularly in mammalian cells. Recently, we demonstrated that cyclin-dependent protein kinases (Cdks) control interphase NPC formation in dividing human cells. Cdks drive the very early step of NPC formation because Cdk inhibition suppressed the generation of "nascent pores," which are considered to be immature NPCs, and disturbed expression and localization of some nucleoporins. Cdk inhibition did not affect nuclear volume, suggesting that these two processes have distinct regulatory mechanisms in the cell cycle. The details of our experimental systems and finding are discussed in more depth. With new findings recently reported, we also discuss possible molecular mechanisms of interphase NPC formation.


Subject(s)
Cell Cycle , Cell Nucleus Size , Nuclear Pore/metabolism , Animals , Cell Cycle/drug effects , Cell Nucleus Size/drug effects , Cricetinae , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans , Interphase/drug effects , Mitosis/drug effects , Neoplasms/pathology , Nuclear Pore/drug effects , Protein Kinase Inhibitors/pharmacology
15.
Cell Physiol Biochem ; 27(5): 471-8, 2011.
Article in English | MEDLINE | ID: mdl-21691064

ABSTRACT

BACKGROUND: NADPH oxidases play an essential role in reactive oxygen species (ROS)-based signaling in the heart. Previously, we have demonstrated that (peri)nuclear expression of the catalytic NADPH oxidase subunit NOX2 in stressed cardiomyocytes, e.g. under ischemia or high concentrations of homocysteine, is an important step in the induction of apoptosis in these cells. Here this ischemia-induced nuclear targeting and activation of NOX2 was specified in cardiomyocytes. METHODS: The effect of ischemia, mimicked by metabolic inhibition, on nuclear localization of NOX2 and the NADPH oxidase subunits p22(phox) and p47(phox), was analyzed in rat neonatal cardiomyoblasts (H9c2 cells) using Western blot, immuno-electron microscopy and digital-imaging microscopy. RESULTS: NOX2 expression significantly increased in nuclear fractions of ischemic H9c2 cells. In addition, in these cells NOX2 was found to colocalize in the nuclear envelope with nuclear pore complexes, p22(phox), p47(phox) and nitrotyrosine residues, a marker for the generation of ROS. Inhibition of NADPH oxidase activity, with apocynin and DPI, significantly reduced (peri)nuclear expression of nitrotyrosine. CONCLUSION: We for the first time show that NOX2, p22(phox) and p47(phox) are targeted to and produce ROS at the nuclear pore complex in ischemic cardiomyocytes.


Subject(s)
Ischemia/pathology , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidases/metabolism , Nuclear Pore/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Acetophenones/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression , Ischemia/chemically induced , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Microscopy, Immunoelectron , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Onium Compounds/pharmacology , Rats , Sodium Cyanide/adverse effects , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
J Cell Mol Med ; 15(4): 928-37, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20497490

ABSTRACT

Mitogen activated protein (MAP) kinases control eukaryotic proliferation, and import of kinases into the nucleus through the nuclear pore complex (NPC) can influence gene expression to affect cellular growth, cell viability and homeostatic function. The NPC is a critical regulatory checkpoint for nucleocytoplasmic traffic that regulates gene expression and cell growth, and MAP kinases may be physically associated with the NPC to modulate transport. In the present study, highly enriched NPC fractions were isolated and investigated for associated kinases and/or activity. Endogenous kinase activity was identified within the NPC fraction, which phosphorylated a 30 kD nuclear pore protein. Phosphomodification of this nucleoporin, here termed Nup30, was inhibited by apigenin and PD-98059, two MAP kinase antagonists as well as with SB-202190, a pharmacological blocker of p38. Furthermore, high throughput profiling of enriched NPCs revealed constitutive presence of all members of the MAP kinase family, extracellular regulated kinases (ERK), p38 and Jun N-terminal kinase. The NPC thus contains a spectrum of associated MAP kinases that suggests an intimate role for ERK and p38 in regulation of nuclear pore function.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Nuclear Pore/enzymology , Animals , Calcium/metabolism , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Nuclear Pore/drug effects , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Substrate Specificity/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Nanomedicine ; 6(5): 605-11, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20470907

ABSTRACT

Advances in nanomedicine require conceptual understanding of physiological processes. Apoptosis is a fundamental physiological process that is characterized, among other things, by an increased permeability of the nuclear envelope (NE). The latter is a tight transport barrier, known to restrict nuclear delivery rate of therapeutic nanoparticles. Therefore, an understanding of the underlying mechanism that leads to the breakdown of the barrier during apoptosis could stimulate the development of new approaches in gene therapy. We set out to elucidate this mechanism following induction of apoptosis on isolated cell nuclei. We tested the hypothesis whether caspases, mediators of apoptosis, trigger the NE leakiness at the level of the nuclear pore complexes (NPCs) using fluorescence techniques. As the permeability barrier inside the NPC channel is thought to be based on hydrophobic-hydrophobic protein interactions we further investigated the NPC channel hydrophobicity using atomic force microscopy. Caspase-9 was found to induce NE leakiness to large macromolecules. Leakiness was prevented by pretreatment of NPCs with an importin-ß mutant, which irreversibly binds and thereby obstructs the NPC channel. Utilizing an ultra-sharp, hydrophobic atomic force microscope tip as a chemical nanosensor that reaches deep into the apoptotic NPC channel, a remarkable decrease of hydrophobic binding sites was detected therein. We conclude that caspase 9 gives rise to NE leakiness by perturbing the hydrophobicity-based barrier inside the NPC channel. This explains the high passive NE permeability in early apoptosis. FROM THE CLINICAL EDITOR: In this study, biological processes taking place in the nucleus during the course of apoptosis have been monitored using atomic force microscopy-based nanosensors. The conclusion was that one of the caspases, caspase 9 perturbs the hydrophobicity-based barrier inside the nuclear pore complex channel causing nuclear envelope leakiness.


Subject(s)
Caspase 9/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Animals , Apoptosis/drug effects , Cytochromes c/pharmacology , Hydrophobic and Hydrophilic Interactions , Microscopy, Atomic Force , Nuclear Envelope/drug effects , Nuclear Pore/drug effects , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Xenopus laevis
18.
Nucleic Acids Res ; 35(12): e86, 2007.
Article in English | MEDLINE | ID: mdl-17584788

ABSTRACT

One of the major obstacles in non-viral gene transfer is the nuclear membrane. Attempts to improve the transport of DNA to the nucleus through the use of nuclear localization signals or importin-beta have achieved limited success. It has been proposed that the nuclear pore complexes (NPCs) through which nucleocytoplasmic transport occurs are filled with a hydrophobic phase through which hydrophobic importins can dissolve. Therefore, considering the hydrophobic nature of the NPC channel, we evaluated whether a non-selective gating of nuclear pores by trans-cyclohexane-1,2-diol (TCHD), an amphipathic alcohol that reversibly collapses the permeability barrier of the NPCs, could be obtained and used as an alternative method to facilitate nuclear entry of plasmid DNA. Our data demonstrate for the first time that TCHD makes the nucleus permeable for both high molecular weight dextrans and plasmid DNA (pDNA) at non-toxic concentrations. Furthermore, in line with these observations, TCHD enhanced the transfection efficacy of both naked DNA and lipoplexes. In conclusion, based on the proposed structure of NPCs we succeeded to temporarily open the NPCs for macromolecules as large as pDNAs and demonstrated that this can significantly enhance non-viral gene delivery.


Subject(s)
Cell Nucleus/metabolism , Cyclohexanols/pharmacology , DNA/metabolism , Nuclear Pore/metabolism , Plasmids/metabolism , Transfection/methods , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Cyclohexanols/chemistry , Cyclohexanols/toxicity , Dextrans/metabolism , Genetic Vectors , Nuclear Pore/drug effects
19.
Bioessays ; 28(9): 935-42, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16937365

ABSTRACT

Nuclear pore complexes mediate and control transport between the cytosol and the nucleus. They form a highly selective and, thus, tight nuclear barrier between these compartments. The nuclear barrier provides the cell with the opportunity to control access to its DNA, a defining feature of eukaryotes. The tightness of the nuclear barrier is therefore physiologically pivotal and any remarkable change in its structure and permeability can prove pathophysiological, e.g. as a result of viral attack. However, there is accumulating evidence that nuclear barrier structure and permeability are highly responsive to hydrophobic cargos of crucial physiological and therapeutic relevance, glucocorticoids (steroid hormones). The present review highlights the glucocorticoid-induced effects on the nuclear barrier structure and permeability concluding that they are physiologically essential to mediate glucocorticoid action.


Subject(s)
Glucocorticoids/pharmacology , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Animals , Genetic Therapy , Hydrophobic and Hydrophilic Interactions , Nuclear Pore/ultrastructure , Nuclear Pore/virology , Viruses/metabolism
20.
Cell Physiol Biochem ; 17(5-6): 181-92, 2006.
Article in English | MEDLINE | ID: mdl-16790994

ABSTRACT

A Glucocorticoid-sensitive cell rapidly responds to hormone stimulation with bidirectional exchange of specific macromolecules between cytosol and nucleus. Glucocorticoid-initiated macromolecules (GIMs) must overcome the nuclear envelope (NE) to enter or leave the nucleus. GIM translocation occurs through nuclear pore complexes (NPCs) that span the NE. We investigated the question whether transport of GIMs through NPCs occurs random or involves selected groups of NPCs (hot spots). Glucocorticoid receptors were expressed in Xenopus laevis oocytes and GIM transport was activated by triamcinolone acetonide, a potent synthetic glucocorticoid analogon. Glucocorticoid receptors associated with the NE and the chromatin were identified using western blot analysis and, at single molecule level, atomic force microscopy. Fluorescence-labeled dextran was used to describe passive NE permeability. We observed that after hormone injection (i) small GIMs, most likely GRs, localize within seconds on both sides of the NE. (ii) large GIMs, most likely ribonucleoproteins, localize within minutes on NPCs at the nucleoplasmic side (iii) both small and large GIMs accumulate on selected NPC clusters (iv) NE permeability transiently decreases when GIMs attach to NPCs. We conclude that GIM transport across the nuclear barrier does not randomly take place but is carried out by a selected population of NPCs.


Subject(s)
Nuclear Envelope/drug effects , Nuclear Pore/metabolism , Oocytes/drug effects , Steroids/pharmacology , Animals , Biological Transport/drug effects , Blotting, Western , Cells, Cultured , Female , Microscopy, Atomic Force , Nuclear Envelope/metabolism , Nuclear Pore/drug effects , Oocytes/cytology , Permeability , Receptors, Glucocorticoid/metabolism , Triamcinolone Acetonide/pharmacology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...